151
|
Yoo T, Cho H, Park H, Lee J, Kim E. Shank3 Exons 14-16 Deletion in Glutamatergic Neurons Leads to Social and Repetitive Behavioral Deficits Associated With Increased Cortical Layer 2/3 Neuronal Excitability. Front Cell Neurosci 2019; 13:458. [PMID: 31649512 PMCID: PMC6795689 DOI: 10.3389/fncel.2019.00458] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 09/26/2019] [Indexed: 12/28/2022] Open
Abstract
Shank3, an abundant excitatory postsynaptic scaffolding protein, has been associated with multiple brain disorders, including autism spectrum disorders (ASD) and Phelan-McDermid syndrome (PMS). However, how cell type-specific Shank3 deletion affects disease-related neuronal and brain functions remains largely unclear. Here, we investigated the impacts of Shank3 deletion in glutamatergic neurons on synaptic and behavioral phenotypes in mice and compared results with those previously obtained from mice with global Shank3 mutation and GABAergic neuron-specific Shank3 mutation. Neuronal excitability was abnormally increased in layer 2/3 pyramidal neurons in the medial prefrontal cortex (mPFC) in mice with a glutamatergic Shank3 deletion, similar to results obtained in mice with a global Shank3 deletion. In addition, excitatory synaptic transmission was abnormally increased in layer 2/3 neurons in mice with a global, but not a glutamatergic, Shank3 deletion, suggesting that Shank3 in glutamatergic neurons are important for the increased neuronal excitability, but not for the increased excitatory synaptic transmission. Neither excitatory nor inhibitory synaptic transmission was altered in the dorsal striatum of Shank3-deficient glutamatergic neurons, a finding that contrasts with the decreased excitatory synaptic transmission in global and Shank3-deficient GABAergic neurons. Behaviorally, glutamatergic Shank3-deficient mice displayed abnormally increased direct social interaction and repetitive self-grooming, similar to global and GABAergic Shank3-deficient mice. These results suggest that glutamatergic and GABAergic Shank3 deletions lead to distinct synaptic and neuronal changes in cortical layer 2/3 and dorsal striatal neurons, but cause similar social and repetitive behavioral abnormalities likely through distinct mechanisms.
Collapse
Affiliation(s)
- Taesun Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Heejin Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea
| | - Haram Park
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Jiseok Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, South Korea.,Center for Synaptic Brain Dysfunctions, Institute for Basic Science (IBS), Daejeon, South Korea
| |
Collapse
|
152
|
Yang G, Shcheglovitov A. Probing disrupted neurodevelopment in autism using human stem cell-derived neurons and organoids: An outlook into future diagnostics and drug development. Dev Dyn 2019; 249:6-33. [PMID: 31398277 DOI: 10.1002/dvdy.100] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 07/23/2019] [Accepted: 07/31/2019] [Indexed: 12/11/2022] Open
Abstract
Autism spectrum disorders (ASDs) represent a spectrum of neurodevelopmental disorders characterized by impaired social interaction, repetitive or restrictive behaviors, and problems with speech. According to a recent report by the Centers for Disease Control and Prevention, one in 68 children in the US is diagnosed with ASDs. Although ASD-related diagnostics and the knowledge of ASD-associated genetic abnormalities have improved in recent years, our understanding of the cellular and molecular pathways disrupted in ASD remains very limited. As a result, no specific therapies or medications are available for individuals with ASDs. In this review, we describe the neurodevelopmental processes that are likely affected in the brains of individuals with ASDs and discuss how patient-specific stem cell-derived neurons and organoids can be used for investigating these processes at the cellular and molecular levels. Finally, we propose a discovery pipeline to be used in the future for identifying the cellular and molecular deficits and developing novel personalized therapies for individuals with idiopathic ASDs.
Collapse
Affiliation(s)
- Guang Yang
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah.,Neuroscience Graduate Program, University of Utah, Salt Lake City, Utah
| | - Alex Shcheglovitov
- Department of Neurobiology and Anatomy, University of Utah, Salt Lake City, Utah.,Neuroscience Graduate Program, University of Utah, Salt Lake City, Utah
| |
Collapse
|
153
|
Shao LX, Jiang Q, Liu XX, Gong DM, Yin YX, Wu G, Sun NH, Wang CK, Chen QZ, Yu C, Shi WX, Fan HY, Fukunaga K, Chen Z, Lu YM, Han F. Functional coupling of Tmem74 and HCN1 channels regulates anxiety-like behavior in BLA neurons. Mol Psychiatry 2019; 24:1461-1477. [PMID: 30886335 DOI: 10.1038/s41380-019-0402-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 02/21/2019] [Accepted: 03/05/2019] [Indexed: 02/08/2023]
Abstract
Anxiety disorders are the most prevalent psychiatric disorders, but their pathogenic mechanism remains poorly understood. Here, we report that transmembrane protein 74 (TMEM74), which contains two putative transmembrane domains and exhibits high levels of mRNA in the brain, is closely associated with the pathogenesis of anxiety disorders. TMEM74 was decreased in the serum of patients with anxiety and the basolateral amygdaloid nucleus (BLA) in chronic stress mice. Furthermore, genetic deletion of Tmem74 or selective knockdown of Tmem74 in BLA pyramidal neurons resulted in anxiety-like behaviors in mice. Whole-cell recordings in BLA pyramidal neurons revealed lower hyperpolarization-activated cation current (Ih) and greater input resistance and excitability in Tmem74-/- neurons than in wild-type neurons. Accordingly, surface expression of hyperpolarization-activated cyclic nucleotide-gated 1 (HCN1) channels was also lower in the BLA of Tmem74-/- mice. The Ih current blocker ZD7288 mimicked these effects in BLA pyramidal neurons in wild-type mice but not in Tmem74-/- mice. Consistent with the improvement in anxiety-like behaviors, Tmem74 overexpression restored HCN1 channel trafficking and pyramidal neuron excitability in the BLA of Tmem74-/- and chronic stress mice. Mechanistically, we demonstrate that interactions between Tmem74 and HCN1 are physiologically relevant and that transmembrane domain 1 (TM1) is essential for the cellular membrane localization of Tmem74 to enhance Ih. Together, our findings suggest that Tmem74 coupling with HCN1 acts as a critical component in the pathophysiology of anxiety and is a potential target for new treatments of anxiety disorders.
Collapse
Affiliation(s)
- Ling-Xiao Shao
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Quan Jiang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiu-Xiu Liu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Dong-Mei Gong
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Yi-Xuan Yin
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.,School of Medicine, Zhejiang University City College, Hangzhou, China
| | - Gang Wu
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Ning-He Sun
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Cheng-Kun Wang
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiao-Zhen Chen
- Department of Psychiatry, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chao Yu
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, China
| | - Wei-Xing Shi
- Departments of Pharmaceutical, Administrative, and Basic Sciences, Schools of Pharmacy and Medicine, Loma Linda University Health, Loma Linda, CA, USA
| | - Heng-Yu Fan
- Life Sciences Institute and Innovation Center for Cell Biology, Zhejiang University, Hangzhou, China
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Zhong Chen
- Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China.
| | - Ying-Mei Lu
- School of Medicine, Zhejiang University City College, Hangzhou, China. .,Department of Neurobiology, Nanjing Medical University, Nanjing, China.
| | - Feng Han
- Key Laboratory of Cardiovascular & Cerebrovascular Medicine, School of Pharmacy, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
154
|
Iakoucheva LM, Muotri AR, Sebat J. Getting to the Cores of Autism. Cell 2019; 178:1287-1298. [PMID: 31491383 PMCID: PMC7039308 DOI: 10.1016/j.cell.2019.07.037] [Citation(s) in RCA: 170] [Impact Index Per Article: 28.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2018] [Revised: 06/07/2019] [Accepted: 07/18/2019] [Indexed: 12/31/2022]
Abstract
The genetic architecture of autism spectrum disorder (ASD) is itself a diverse allelic spectrum that consists of rare de novo or inherited variants in hundreds of genes and common polygenic risk at thousands of loci. ASD susceptibility genes are interconnected at the level of transcriptional and protein networks, and many function as genetic regulators of neurodevelopment or synaptic proteins that regulate neural activity. So that the core underlying neuropathologies can be further elucidated, we emphasize the importance of first defining subtypes of ASD on the basis of the phenotypic signatures of genes in model systems and humans.
Collapse
Affiliation(s)
- Lilia M Iakoucheva
- University of California San Diego, Department of Psychiatry, La Jolla, CA 92093, USA
| | - Alysson R Muotri
- University of California San Diego, School of Medicine, Department of Cellular & Molecular Medicine, La Jolla, CA 92093, USA; University of California San Diego, School of Medicine, Department of Pediatrics/Rady Children's Hospital San Diego, La Jolla, CA 92093, USA; University of California San Diego, Kavli Institute for Brain and Mind, La Jolla, CA 92093, USA; Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA 92093, USA
| | - Jonathan Sebat
- University of California San Diego, Department of Psychiatry, La Jolla, CA 92093, USA; University of California San Diego, School of Medicine, Department of Cellular & Molecular Medicine, La Jolla, CA 92093, USA; University of California San Diego, Beyster Center for Psychiatric Genomics, La Jolla, CA 92093.
| |
Collapse
|
155
|
Flitsch LJ, Brüstle O. Evolving principles underlying neural lineage conversion and their relevance for biomedical translation. F1000Res 2019; 8. [PMID: 31559012 PMCID: PMC6743253 DOI: 10.12688/f1000research.18926.1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2019] [Indexed: 12/19/2022] Open
Abstract
Scientific and technological advances of the past decade have shed light on the mechanisms underlying cell fate acquisition, including its transcriptional and epigenetic regulation during embryonic development. This knowledge has enabled us to purposefully engineer cell fates
in vitro by manipulating expression levels of lineage-instructing transcription factors. Here, we review the state of the art in the cell programming field with a focus on the derivation of neural cells. We reflect on what we know about the mechanisms underlying fate changes in general and on the degree of epigenetic remodeling conveyed by the distinct reprogramming and direct conversion strategies available. Moreover, we discuss the implications of residual epigenetic memory for biomedical applications such as disease modeling and neuroregeneration. Finally, we cover recent developments approaching cell fate conversion in the living brain and define questions which need to be addressed before cell programming can become an integral part of translational medicine.
Collapse
Affiliation(s)
- Lea Jessica Flitsch
- Institute of Reconstructive Neurobiology, University of Bonn School of Medicine & University Hospital Bonn, Bonn, North Rhine Wesphalia, 53127, Germany
| | - Oliver Brüstle
- Institute of Reconstructive Neurobiology, University of Bonn School of Medicine & University Hospital Bonn, Bonn, North Rhine Wesphalia, 53127, Germany
| |
Collapse
|
156
|
Abstract
The structure of neuronal circuits that subserve cognitive functions in the brain is shaped and refined throughout development and into adulthood. Evidence from human and animal studies suggests that the cellular and synaptic substrates of these circuits are atypical in neuropsychiatric disorders, indicating that altered structural plasticity may be an important part of the disease biology. Advances in genetics have redefined our understanding of neuropsychiatric disorders and have revealed a spectrum of risk factors that impact pathways known to influence structural plasticity. In this Review, we discuss the importance of recent genetic findings on the different mechanisms of structural plasticity and propose that these converge on shared pathways that can be targeted with novel therapeutics.
Collapse
|
157
|
Orefice LL, Mosko JR, Morency DT, Wells MF, Tasnim A, Mozeika SM, Ye M, Chirila AM, Emanuel AJ, Rankin G, Fame RM, Lehtinen MK, Feng G, Ginty DD. Targeting Peripheral Somatosensory Neurons to Improve Tactile-Related Phenotypes in ASD Models. Cell 2019; 178:867-886.e24. [PMID: 31398341 PMCID: PMC6704376 DOI: 10.1016/j.cell.2019.07.024] [Citation(s) in RCA: 159] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/06/2019] [Accepted: 07/11/2019] [Indexed: 12/23/2022]
Abstract
Somatosensory over-reactivity is common among patients with autism spectrum disorders (ASDs) and is hypothesized to contribute to core ASD behaviors. However, effective treatments for sensory over-reactivity and ASDs are lacking. We found distinct somatosensory neuron pathophysiological mechanisms underlie tactile abnormalities in different ASD mouse models and contribute to some ASD-related behaviors. Developmental loss of ASD-associated genes Shank3 or Mecp2 in peripheral mechanosensory neurons leads to region-specific brain abnormalities, revealing links between developmental somatosensory over-reactivity and the genesis of aberrant behaviors. Moreover, acute treatment with a peripherally restricted GABAA receptor agonist that acts directly on mechanosensory neurons reduced tactile over-reactivity in six distinct ASD models. Chronic treatment of Mecp2 and Shank3 mutant mice improved body condition, some brain abnormalities, anxiety-like behaviors, and some social impairments but not memory impairments, motor deficits, or overgrooming. Our findings reveal a potential therapeutic strategy targeting peripheral mechanosensory neurons to treat tactile over-reactivity and select ASD-related behaviors.
Collapse
Affiliation(s)
- Lauren L Orefice
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Jacqueline R Mosko
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Danielle T Morency
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Michael F Wells
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge, MA 02139, USA
| | - Aniqa Tasnim
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Shawn M Mozeika
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Mengchen Ye
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Anda M Chirila
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Alan J Emanuel
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Genelle Rankin
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA
| | - Ryann M Fame
- Department of Pathology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Guoping Feng
- McGovern Institute for Brain Research, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, 43 Vassar Street, Cambridge, MA 02139, USA
| | - David D Ginty
- Department of Neurobiology, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA; Howard Hughes Medical Institute, Harvard Medical School, 220 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
158
|
Differential Signaling Mediated by ApoE2, ApoE3, and ApoE4 in Human Neurons Parallels Alzheimer's Disease Risk. J Neurosci 2019; 39:7408-7427. [PMID: 31331998 DOI: 10.1523/jneurosci.2994-18.2019] [Citation(s) in RCA: 97] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 07/09/2019] [Accepted: 07/12/2019] [Indexed: 12/25/2022] Open
Abstract
In blood, apolipoprotein E (ApoE) is a component of circulating lipoproteins and mediates the clearance of these lipoproteins from blood by binding to ApoE receptors. Humans express three genetic ApoE variants, ApoE2, ApoE3, and ApoE4, which exhibit distinct ApoE receptor-binding properties and differentially affect Alzheimer's disease (AD), such that ApoE2 protects against, and ApoE4 predisposes to AD. In brain, ApoE-containing lipoproteins are secreted by activated astrocytes and microglia, but their functions and role in AD pathogenesis are largely unknown. Ample evidence suggests that ApoE4 induces microglial dysregulation and impedes Aβ clearance in AD, but the direct neuronal effects of ApoE variants are poorly studied. Extending previous studies, we here demonstrate that the three ApoE variants differentially activate multiple neuronal signaling pathways and regulate synaptogenesis. Specifically, using human neurons (male embryonic stem cell-derived) cultured in the absence of glia to exclude indirect glial mechanisms, we show that ApoE broadly stimulates signal transduction cascades. Among others, such stimulation enhances APP synthesis and synapse formation with an ApoE4>ApoE3>ApoE2 potency rank order, paralleling the relative risk for AD conferred by these ApoE variants. Unlike the previously described induction of APP transcription, however, ApoE-induced synaptogenesis involves CREB activation rather than cFos activation. We thus propose that in brain, ApoE acts as a glia-secreted signal that activates neuronal signaling pathways. The parallel potency rank order of ApoE4>ApoE3>ApoE2 in AD risk and neuronal signaling suggests that ApoE4 may in an apparent paradox promote AD pathogenesis by causing a chronic increase in signaling, possibly via enhancing APP expression.SIGNIFICANCE STATEMENT Humans express three genetic variants of apolipoprotein E (ApoE), ApoE2, ApoE3, and ApoE4. ApoE4 constitutes the most important genetic risk factor for Alzheimer's disease (AD), whereas ApoE2 protects against AD. Significant evidence suggests that ApoE4 impairs microglial function and impedes astrocytic Aβ clearance in brain, but the direct neuronal effects of ApoE are poorly understood, and the differences between ApoE variants in these effects are unclear. Here, we report that ApoE acts on neurons as a glia-secreted signaling molecule that, among others, enhances synapse formation. In activating neuronal signaling, the three ApoE variants exhibit a differential potency of ApoE4>ApoE3>ApoE2, which mirrors their relative effects on AD risk, suggesting that differential signaling by ApoE variants may contribute to AD pathogenesis.
Collapse
|
159
|
Marini C, Porro A, Rastetter A, Dalle C, Rivolta I, Bauer D, Oegema R, Nava C, Parrini E, Mei D, Mercer C, Dhamija R, Chambers C, Coubes C, Thévenon J, Kuentz P, Julia S, Pasquier L, Dubourg C, Carré W, Rosati A, Melani F, Pisano T, Giardino M, Innes AM, Alembik Y, Scheidecker S, Santos M, Figueiroa S, Garrido C, Fusco C, Frattini D, Spagnoli C, Binda A, Granata T, Ragona F, Freri E, Franceschetti S, Canafoglia L, Castellotti B, Gellera C, Milanesi R, Mancardi MM, Clark DR, Kok F, Helbig KL, Ichikawa S, Sadler L, Neupauerová J, Laššuthova P, Šterbová K, Laridon A, Brilstra E, Koeleman B, Lemke JR, Zara F, Striano P, Soblet J, Smits G, Deconinck N, Barbuti A, DiFrancesco D, LeGuern E, Guerrini R, Santoro B, Hamacher K, Thiel G, Moroni A, DiFrancesco JC, Depienne C. HCN1 mutation spectrum: from neonatal epileptic encephalopathy to benign generalized epilepsy and beyond. Brain 2019; 141:3160-3178. [PMID: 30351409 DOI: 10.1093/brain/awy263] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Accepted: 08/09/2018] [Indexed: 12/15/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels control neuronal excitability and their dysfunction has been linked to epileptogenesis but few individuals with neurological disorders related to variants altering HCN channels have been reported so far. In 2014, we described five individuals with epileptic encephalopathy due to de novo HCN1 variants. To delineate HCN1-related disorders and investigate genotype-phenotype correlations further, we assembled a cohort of 33 unpublished patients with novel pathogenic or likely pathogenic variants: 19 probands carrying 14 different de novo mutations and four families with dominantly inherited variants segregating with epilepsy in 14 individuals, but not penetrant in six additional individuals. Sporadic patients had epilepsy with median onset at age 7 months and in 36% the first seizure occurred during a febrile illness. Overall, considering familial and sporadic patients, the predominant phenotypes were mild, including genetic generalized epilepsies and genetic epilepsy with febrile seizures plus (GEFS+) spectrum. About 20% manifested neonatal/infantile onset otherwise unclassified epileptic encephalopathy. The study also included eight patients with variants of unknown significance: one adopted patient had two HCN1 variants, four probands had intellectual disability without seizures, and three individuals had missense variants inherited from an asymptomatic parent. Of the 18 novel pathogenic missense variants identified, 12 were associated with severe phenotypes and clustered within or close to transmembrane domains, while variants segregating with milder phenotypes were located outside transmembrane domains, in the intracellular N- and C-terminal parts of the channel. Five recurrent variants were associated with similar phenotypes. Using whole-cell patch-clamp, we showed that the impact of 12 selected variants ranged from complete loss-of-function to significant shifts in activation kinetics and/or voltage dependence. Functional analysis of three different substitutions altering Gly391 revealed that these variants had different consequences on channel biophysical properties. The Gly391Asp variant, associated with the most severe, neonatal phenotype, also had the most severe impact on channel function. Molecular dynamics simulation on channel structure showed that homotetramers were not conducting ions because the permeation path was blocked by cation(s) strongly complexed to the Asp residue, whereas heterotetramers showed an instantaneous current component possibly linked to deformation of the channel pore. In conclusion, our results considerably expand the clinical spectrum related to HCN1 variants to include common generalized epilepsy phenotypes and further illustrate how HCN1 has a pivotal function in brain development and control of neuronal excitability.
Collapse
Affiliation(s)
- Carla Marini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy.,EuroEPINOMICS RES Consortium
| | | | - Agnès Rastetter
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Carine Dalle
- Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France
| | - Ilaria Rivolta
- School of Medicine and Surgery, University Milano-Bicocca, Monza, Italy
| | - Daniel Bauer
- Computational Biology and Simulation Group, Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Renske Oegema
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Caroline Nava
- EuroEPINOMICS RES Consortium.,Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Elena Parrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - Davide Mei
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - Catherine Mercer
- Wessex Clinical Genetics Service, Princess Anne Hospital, Southampton, UK
| | - Radhika Dhamija
- Department of Clinical Genomics and Neurology, Mayo Clinic, Phoenix, AZ, USA
| | - Chelsea Chambers
- Department of Neurosciences, University of Virginia, Charlottesville, VA, USA
| | - Christine Coubes
- Département de Génétique Médicale, Maladies Rares et Médecine Personnalisée, Hôpital Arnaud de Villeneuve, Montpellier, France
| | - Julien Thévenon
- FHU-TRANSLAD, Université de Bourgogne/CHU Dijon and INSERM UMR 1231 GAD team, Genetics of Developmental Anomalies, Université de Bourgogne-Franche Comté, Dijon, France
| | - Paul Kuentz
- FHU-TRANSLAD, Université de Bourgogne/CHU Dijon and INSERM UMR 1231 GAD team, Genetics of Developmental Anomalies, Université de Bourgogne-Franche Comté, Dijon, France.,Génétique Biologique Histologie, CHRU de Besançon, Besançon, France
| | - Sophie Julia
- Service de génétique médicale, Pôle de biologie, CHU de Toulouse - Hôpital Purpan, Toulouse, France
| | - Laurent Pasquier
- Service de Génétique Clinique, Centre Référence Déficiences Intellectuelles de causes rares (CRDI), CHU Rennes, Rennes, France
| | - Christèle Dubourg
- Laboratoire de Génétique Moléculaire et Génomique, CHU de Rennes, Rennes, France
| | - Wilfrid Carré
- Laboratoire de Génétique Moléculaire et Génomique, CHU de Rennes, Rennes, France
| | - Anna Rosati
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - Federico Melani
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - Tiziana Pisano
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - Maria Giardino
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy
| | - A Micheil Innes
- Department of Medical Genetics and Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Yves Alembik
- Laboratoires de génétique, Institut de génétique médicale d'Alsace, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Sophie Scheidecker
- Laboratoires de génétique, Institut de génétique médicale d'Alsace, Hôpitaux Universitaires de Strasbourg, Strasbourg, France
| | - Manuela Santos
- Neuropediatric Department, Centro Hospitalar do Porto, Porto, Portugal
| | - Sonia Figueiroa
- Neuropediatric Department, Centro Hospitalar do Porto, Porto, Portugal
| | - Cristina Garrido
- Neuropediatric Department, Centro Hospitalar do Porto, Porto, Portugal
| | - Carlo Fusco
- Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Daniele Frattini
- Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Carlotta Spagnoli
- Azienda Unità Sanitaria Locale - IRCCS di Reggio Emilia, Reggio Emilia, Italy
| | - Anna Binda
- School of Medicine and Surgery, University Milano-Bicocca, Monza, Italy
| | - Tiziana Granata
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | - Elena Freri
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | | | | | | | - Cinzia Gellera
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Raffaella Milanesi
- Department of Biosciences, The PaceLab, Università degli Studi di Milano, Milan, Italy
| | - Maria Margherita Mancardi
- Child Neuropsychiatry Unit, Department of Medical and Surgical Neurosciences and Rehabilitation, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | | | - Fernando Kok
- Mendelics Genomic Analysis, Sao Paulo, SP, Brazil
| | - Katherine L Helbig
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Shoji Ichikawa
- Department of Clinical Diagnostics, Ambry Genetics, Aliso Viejo, CA, USA
| | - Laurie Sadler
- Division of Genetics, Department of Pediatrics, Oishei Children's Hospital, Jacobs School of Medicine and Biomedical Sciences, University of Buffalo, State University of New York, Buffalo, NY, USA
| | - Jana Neupauerová
- Department of Child Neurology, Charles University 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Petra Laššuthova
- Department of Child Neurology, Charles University 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Katalin Šterbová
- EuroEPINOMICS RES Consortium.,Department of Child Neurology, Charles University 2nd Faculty of Medicine and University Hospital Motol, Prague, Czech Republic
| | - Annick Laridon
- Department of Neurology, Academic Center for Epileptology, Kempenhaeghe/Maastricht University Medical Center, Heeze, The Netherlands
| | - Eva Brilstra
- EuroEPINOMICS RES Consortium.,Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Bobby Koeleman
- EuroEPINOMICS RES Consortium.,Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Johannes R Lemke
- EuroEPINOMICS RES Consortium.,Institute of Human Genetics, University of Leipzig Hospitals and Clinics, Leipzig, Germany
| | - Federico Zara
- Laboratory of Neurogenetics and Neuroscience, Institute G Gaslini, Genova, Italy
| | - Pasquale Striano
- EuroEPINOMICS RES Consortium.,Pediatric Neurology and Muscular Diseases Unit, Department of Neurosciences, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health, University of Genoa, 'G Gaslini' Institute, Genova, Italy
| | - Julie Soblet
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Department of Genetics, Hôpital Erasme ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles, Brussels, Belgium
| | - Guillaume Smits
- Department of Genetics, Hôpital Universitaire des Enfants Reine Fabiola, ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Department of Genetics, Hôpital Erasme ULB Center of Human Genetics, Université Libre de Bruxelles, Brussels, Belgium.,Interuniversity Institute of Bioinformatics in Brussels, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Deconinck
- Department of Pediatric Neurology, Hôpital Universitaire des Enfants Reine Fabiola, Université Libre de Bruxelles, ULB, Brussels, Belgium
| | - Andrea Barbuti
- Department of Biosciences, The PaceLab, Università degli Studi di Milano, Milan, Italy
| | - Dario DiFrancesco
- Department of Biosciences, The PaceLab, Università degli Studi di Milano, Milan, Italy
| | - Eric LeGuern
- EuroEPINOMICS RES Consortium.,Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,AP-HP, Groupe Hospitalier Pitié-Salpêtrière, Département de Génétique, Paris, France
| | - Renzo Guerrini
- Pediatric Neurology, Neurogenetics and Neurobiology Unit and Laboratories, Neuroscience Department, A Meyer Children's Hospital, University of Florence, Viale Pieraccini 24, Florence, Italy.,EuroEPINOMICS RES Consortium
| | - Bina Santoro
- Department of Neuroscience, Columbia University, New York, NY, USA
| | - Kay Hamacher
- Computational Biology and Simulation Group, Department of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Gerhard Thiel
- Membrane Biophysics, Deparment of Biology, Technische Universität Darmstadt, Darmstadt, Germany
| | - Anna Moroni
- Department of Biosciences, University of Milan, Milan, Italy
| | - Jacopo C DiFrancesco
- Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy.,Department of Neurology, San Gerardo Hospital, University Milano-Bicocca, Monza, Italy
| | - Christel Depienne
- EuroEPINOMICS RES Consortium.,Inserm U 1127, CNRS UMR 7225, Sorbonne Universités, UPMC Univ Paris 06 UMR S 1127, Institut du Cerveau et de la Moelle épinière, ICM, Paris, France.,IGBMC, CNRS UMR 7104/INSERM U964/Université de Strasbourg, Illkirch, France.,Institute of Human Genetics, University Hospital Essen, University Duisburg-Essen, Essen, Germany
| |
Collapse
|
160
|
Di Nanni N, Bersanelli M, Cupaioli FA, Milanesi L, Mezzelani A, Mosca E. Network-Based Integrative Analysis of Genomics, Epigenomics and Transcriptomics in Autism Spectrum Disorders. Int J Mol Sci 2019; 20:E3363. [PMID: 31323926 PMCID: PMC6651137 DOI: 10.3390/ijms20133363] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 07/05/2019] [Accepted: 07/06/2019] [Indexed: 01/16/2023] Open
Abstract
Current studies suggest that autism spectrum disorders (ASDs) may be caused by many genetic factors. In fact, collectively considering multiple studies aimed at characterizing the basic pathophysiology of ASDs, a large number of genes has been proposed. Addressing the problem of molecular data interpretation using gene networks helps to explain genetic heterogeneity in terms of shared pathways. Besides, the integrative analysis of multiple omics has emerged as an approach to provide a more comprehensive view of a disease. In this work, we carry out a network-based meta-analysis of the genes reported as associated with ASDs by studies that involved genomics, epigenomics, and transcriptomics. Collectively, our analysis provides a prioritization of the large number of genes proposed to be associated with ASDs, based on genes' relevance within the intracellular circuits, the strength of the supporting evidence of association with ASDs, and the number of different molecular alterations affecting genes. We discuss the presence of the prioritized genes in the SFARI (Simons Foundation Autism Research Initiative) database and in gene networks associated with ASDs by other investigations. Lastly, we provide the full results of our analyses to encourage further studies on common targets amenable to therapy.
Collapse
Affiliation(s)
- Noemi Di Nanni
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate (MI), Italy
- Department of Industrial and Information Engineering, University of Pavia, Via Ferrata 5, 27100 Pavia, Italy
| | - Matteo Bersanelli
- Department of Physics and Astronomy, University of Bologna, Via B. Pichat 6/2, 40127 Bologna, Italy
- National Institute of Nuclear Physics (INFN), 40127 Bologna, Italy
| | - Francesca Anna Cupaioli
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate (MI), Italy
| | - Luciano Milanesi
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate (MI), Italy
| | - Alessandra Mezzelani
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate (MI), Italy
| | - Ettore Mosca
- Institute of Biomedical Technologies, Italian National Research Council, Via Fratelli Cervi 93, 20090 Segrate (MI), Italy.
| |
Collapse
|
161
|
Marro SG, Chanda S, Yang N, Janas JA, Valperga G, Trotter J, Zhou B, Merrill S, Yousif I, Shelby H, Vogel H, Kalani MYS, Südhof TC, Wernig M. Neuroligin-4 Regulates Excitatory Synaptic Transmission in Human Neurons. Neuron 2019; 103:617-626.e6. [PMID: 31257103 DOI: 10.1016/j.neuron.2019.05.043] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 04/17/2019] [Accepted: 05/28/2019] [Indexed: 12/26/2022]
Abstract
The autism-associated synaptic-adhesion gene Neuroligin-4 (NLGN4) is poorly conserved evolutionarily, limiting conclusions from Nlgn4 mouse models for human cells. Here, we show that the cellular and subcellular expression of human and murine Neuroligin-4 differ, with human Neuroligin-4 primarily expressed in cerebral cortex and localized to excitatory synapses. Overexpression of NLGN4 in human embryonic stem cell-derived neurons resulted in an increase in excitatory synapse numbers but a remarkable decrease in synaptic strength. Human neurons carrying the syndromic autism mutation NLGN4-R704C also formed more excitatory synapses but with increased functional synaptic transmission due to a postsynaptic mechanism, while genetic loss of NLGN4 did not significantly affect synapses in the human neurons analyzed. Thus, the NLGN4-R704C mutation represents a change-of-function mutation. Our work reveals contrasting roles of NLGN4 in human and mouse neurons, suggesting that human evolution has impacted even fundamental cell biological processes generally assumed to be highly conserved.
Collapse
Affiliation(s)
- Samuele G Marro
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Soham Chanda
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nan Yang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Justyna A Janas
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Giulio Valperga
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Justin Trotter
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Bo Zhou
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sean Merrill
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Issa Yousif
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hannah Shelby
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Hannes Vogel
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - M Yashar S Kalani
- Departments of Neurosurgery and Neuroscience, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Thomas C Südhof
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA 94305, USA.
| | - Marius Wernig
- Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
162
|
The Autism-Associated Gene Scn2a Contributes to Dendritic Excitability and Synaptic Function in the Prefrontal Cortex. Neuron 2019; 103:673-685.e5. [PMID: 31230762 DOI: 10.1016/j.neuron.2019.05.037] [Citation(s) in RCA: 139] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 03/23/2019] [Accepted: 05/22/2019] [Indexed: 12/20/2022]
Abstract
Autism spectrum disorder (ASD) is strongly associated with de novo gene mutations. One of the most commonly affected genes is SCN2A. ASD-associated SCN2A mutations impair the encoded protein NaV1.2, a sodium channel important for action potential initiation and propagation in developing excitatory cortical neurons. The link between an axonal sodium channel and ASD, a disorder typically attributed to synaptic or transcriptional dysfunction, is unclear. Here we show that NaV1.2 is unexpectedly critical for dendritic excitability and synaptic function in mature pyramidal neurons in addition to regulating early developmental axonal excitability. NaV1.2 loss reduced action potential backpropagation into dendrites, impairing synaptic plasticity and synaptic strength, even when NaV1.2 expression was disrupted in a cell-autonomous fashion late in development. These results reveal a novel dendritic function for NaV1.2, providing insight into cellular mechanisms probably underlying circuit and behavioral dysfunction in ASD.
Collapse
|
163
|
Yoo YE, Yoo T, Lee S, Lee J, Kim D, Han HM, Bae YC, Kim E. Shank3 Mice Carrying the Human Q321R Mutation Display Enhanced Self-Grooming, Abnormal Electroencephalogram Patterns, and Suppressed Neuronal Excitability and Seizure Susceptibility. Front Mol Neurosci 2019; 12:155. [PMID: 31275112 PMCID: PMC6591539 DOI: 10.3389/fnmol.2019.00155] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2018] [Accepted: 06/03/2019] [Indexed: 11/13/2022] Open
Abstract
Shank3, a postsynaptic scaffolding protein involved in regulating excitatory synapse assembly and function, has been implicated in several brain disorders, including autism spectrum disorders (ASD), Phelan-McDermid syndrome, schizophrenia, intellectual disability, and mania. Here we generated and characterized a Shank3 knock-in mouse line carrying the Q321R mutation (Shank3 Q321R mice) identified in a human individual with ASD that affects the ankyrin repeat region (ARR) domain of the Shank3 protein. Homozygous Shank3 Q321R/Q321R mice show a selective decrease in the level of Shank3a, an ARR-containing protein variant, but not other variants. CA1 pyramidal neurons in the Shank3 Q321R/Q321R hippocampus show decreased neuronal excitability but normal excitatory and inhibitory synaptic transmission. Behaviorally, Shank3 Q321R/Q321R mice show moderately enhanced self-grooming and anxiolytic-like behavior, but normal locomotion, social interaction, and object recognition and contextual fear memory. In addition, these mice show abnormal electroencephalogram (EEG) patterns and decreased susceptibility to induced seizures. These results indicate that the Q321R mutation alters Shank3 protein stability, neuronal excitability, repetitive and anxiety-like behavior, EEG patterns, and seizure susceptibility in mice.
Collapse
Affiliation(s)
- Ye-Eun Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Taesun Yoo
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Seungjoon Lee
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea
| | - Jiseok Lee
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| | - Doyoun Kim
- Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| | - Hye-Min Han
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Yong-Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Eunjoon Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology, Daejeon, South Korea.,Center for Synaptic Brain Dysfunctions, Institute for Basic Science, Daejeon, South Korea
| |
Collapse
|
164
|
Chen SJ, Xu Y, Liang YM, Cao Y, Lv JY, Pang JX, Zhou PZ. Identification and characterization of a series of novel HCN channel inhibitors. Acta Pharmacol Sin 2019; 40:746-754. [PMID: 30315249 DOI: 10.1038/s41401-018-0162-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 08/13/2018] [Indexed: 11/09/2022]
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels play a critical role in controlling pacemaker activity in both heart and nervous system. Developing HCN channel inhibitors has been proposed to be an important strategy for the treatment of pain, heart failure, arrhythmias, and epilepsy. One HCN channel inhibitor, ivabradine, has been clinically approved for the treatment of angina pectoris and heart failure. In this study, we designed and synthesized eight alkanol amine derivatives, and assessed their effects on HCN channels expressed in COS7 cells using a whole-cell patch clamp method. Among them, compound 4e displayed the most potent inhibitory activity with an IC50 of 2.9 ± 1.2 µM at - 120 mV on HCN2 channel expressed in COS7 cells. Further analysis revealed that application of compound 4e (10 μM) caused a slowing of activation and a hyperpolarizing shift (ΔV1/2 = - 30.2 ± 2.9 mV, n = 5) in the voltage dependence of HCN2 channel activation. The inhibitory effect of compound 4e on HCN1 and HCN4 channel expressed in COS7 cells was less potent with IC50 of 17.2 ± 1.3 and 7.3 ± 1.2 μM, respectively. Besides, we showed that application of compound 4e (10 μM) inhibited Ih and action potential firing in acutely dissociated mouse small dorsal root ganglion neurons. Our study provides a new strategy for the design and development of potent HCN channel inhibitors.
Collapse
|
165
|
Direct Reprogramming of Human Neurons Identifies MARCKSL1 as a Pathogenic Mediator of Valproic Acid-Induced Teratogenicity. Cell Stem Cell 2019; 25:103-119.e6. [PMID: 31155484 DOI: 10.1016/j.stem.2019.04.021] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 02/19/2019] [Accepted: 04/26/2019] [Indexed: 12/24/2022]
Abstract
Human pluripotent stem cells can be rapidly converted into functional neurons by ectopic expression of proneural transcription factors. Here we show that directly reprogrammed neurons, despite their rapid maturation kinetics, can model teratogenic mechanisms that specifically affect early neurodevelopment. We delineated distinct phases of in vitro maturation during reprogramming of human neurons and assessed the cellular phenotypes of valproic acid (VPA), a teratogenic drug. VPA exposure caused chronic impairment of dendritic morphology and functional properties of developing neurons, but not those of mature neurons. These pathogenic effects were associated with VPA-mediated inhibition of the histone deacetylase (HDAC) and glycogen synthase kinase-3 (GSK-3) pathways, which caused transcriptional downregulation of many genes, including MARCKSL1, an actin-stabilizing protein essential for dendritic morphogenesis and synapse maturation during early neurodevelopment. Our findings identify a developmentally restricted pathogenic mechanism of VPA and establish the use of reprogrammed neurons as an effective platform for modeling teratogenic pathways.
Collapse
|
166
|
Hajisoltani R, Karimi SA, Rahdar M, Davoudi S, Borjkhani M, Hosseinmardi N, Behzadi G, Janahmadi M. Hyperexcitability of hippocampal CA1 pyramidal neurons in male offspring of a rat model of autism spectrum disorder (ASD) induced by prenatal exposure to valproic acid: A possible involvement of Ih channel current. Brain Res 2019; 1708:188-199. [DOI: 10.1016/j.brainres.2018.12.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/30/2018] [Accepted: 12/07/2018] [Indexed: 11/25/2022]
|
167
|
RNA-Sequencing and Bioinformatics Analysis of Long Noncoding RNAs and mRNAs in the Prefrontal Cortex of Mice Following Repeated Social Defeat Stress. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7505260. [PMID: 31032362 PMCID: PMC6457290 DOI: 10.1155/2019/7505260] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/01/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023]
Abstract
Background Repeated or continuous chronic psychological stress may induce diverse neuropsychiatric disorders; however, the underlying mechanisms remain unclear. In this study, we explored the expression profiles of long noncoding RNAs (lncRNAs) and mRNAs, along with their biological function and regulatory network, in mice after repeated social defeat (RSD) stress to explore their potential involvement in the development of anxiety-like behaviors. Main Methods RNA-sequencing was used to screen all differentially expressed (DE) lncRNAs and mRNAs between the RSD and control groups. Quantitative real-time polymerase chain reaction (qRT-PCR) was used for confirmation of the RNA-sequencing results. The function of DE lncRNAs was predicted by Gene Ontology (GO) enrichment and pathway analyses of target mRNAs. In addition, the functional regulatory network of the target mRNAs was constructed to reveal potential relationships between lncRNAs and their target genes with bioinformatics approaches. Key Findings In mice experiencing RSD, 373 and 454 lncRNAs, along with 1142 and 654, mRNAs were significantly upregulated and downregulated, respectively. The detailed regulatory network included 126 eligible lncRNA-mRNA pairs. Among them, 14 genes such as Arhgef1, Chchd2, Fam107a, Dlg1, Nova2, Dpf1, and Shank3 involved in neurite growth, neural development, and synaptic plasticity were direct targets of the DE lncRNAs. qRT-PCR of four of the DE lncRNAs and mRNAs confirmed the reliability of RNA-sequencing. GO clustering analyses showed that the top enriched biological process, cellular component, and molecular function terms were synaptic transmission, neuron spine, and glutamate receptor binding, respectively. Further, the top three significant enriched pathways were synaptic adhesion-like molecule (SALM) protein interactions at the synapses, trafficking of α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors, as well as glutamate binding, activation of AMPA receptors, and synaptic plasticity. Significance Hundreds of lncRNAs and mRNAs are dysregulated after RSD, and many of these lncRNAs might participate in the development of anxiety-like behaviors via multiple complex mechanisms such as target regulation. Available informatics evidence highlighted the likely role of synapse dysfunction and abnormal synaptic neurotransmission in these behaviors. Thus, our findings provide potential candidate biomarkers or intervention targets for chronic psychological stress-induced neuropsychiatric disorders.
Collapse
|
168
|
SHANK2 mutations associated with autism spectrum disorder cause hyperconnectivity of human neurons. Nat Neurosci 2019; 22:556-564. [PMID: 30911184 PMCID: PMC6475597 DOI: 10.1038/s41593-019-0365-8] [Citation(s) in RCA: 115] [Impact Index Per Article: 19.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Accepted: 02/15/2019] [Indexed: 12/13/2022]
Abstract
Heterozygous loss-of-function mutations in SHANK2 are associated with autism spectrum disorder (ASD). We generated cortical neurons from induced pluripotent stem cells (iPSC) derived from neurotypic and ASD-affected donors. We developed Sparse coculture for Connectivity (SparCon) assays where SHANK2 and control neurons were differentially labeled and sparsely seeded together on a lawn of unlabeled control neurons. We observed increases in dendrite length, dendrite complexity, synapse number, and frequency of spontaneous excitatory postsynaptic currents. These findings were phenocopied in gene-edited homozygous SHANK2 knockout cells and rescued by gene correction of an ASD SHANK2 mutation. Dendrite length increases were exacerbated by IGF1, TG003, or BDNF, and suppressed by DHPG treatment. The transcriptome in isogenic SHANK2 neurons was perturbed in synapse, plasticity, and neuronal morphogenesis gene sets and ASD gene modules, and activity-dependent dendrite extension was impaired. Our findings provide evidence for hyperconnectivity and altered transcriptome in SHANK2 neurons derived from ASD subjects.
Collapse
|
169
|
Fenske P, Grauel MK, Brockmann MM, Dorrn AL, Trimbuch T, Rosenmund C. Autaptic cultures of human induced neurons as a versatile platform for studying synaptic function and neuronal morphology. Sci Rep 2019; 9:4890. [PMID: 30894602 PMCID: PMC6427022 DOI: 10.1038/s41598-019-41259-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 03/05/2019] [Indexed: 02/08/2023] Open
Abstract
Recently developed technology to differentiate induced pluripotent stem cells (iPSCs) into human induced neurons (iNs) provides an exciting opportunity to study the function of human neurons. However, functional characterisations of iNs have been hampered by the reliance on mass culturing protocols which do not allow assessment of synaptic release characteristics and neuronal morphology at the individual cell level with quantitative precision. Here, we have developed for the first time a protocol to generate autaptic cultures of iPSC-derived iNs. We show that our method efficiently generates mature, autaptic iNs with robust spontaneous and action potential-driven synaptic transmission. The synaptic responses are sensitive to modulation by metabotropic receptor agonists as well as potentiation by acute phorbol ester application. Finally, we demonstrate loss of evoked and spontaneous release by Unc13A knockdown. This culture system provides a versatile platform allowing for quantitative and integrative assessment of morphophysiological and molecular parameters underlying human synaptic transmission.
Collapse
Affiliation(s)
- Pascal Fenske
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - M Katharina Grauel
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Marisa M Brockmann
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Anja L Dorrn
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Thorsten Trimbuch
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany.,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany
| | - Christian Rosenmund
- Institute of Neurophysiology, Charité - Universitätsmedizin, 10117, Berlin, Germany. .,NeuroCure Cluster of Excellence, Charité - Universitätsmedizin, 10117, Berlin, Germany. .,Berlin Institute of Health, Anna-Louise-Karsch-Straße 2, 10178, Berlin, Germany.
| |
Collapse
|
170
|
Koppensteiner P, Galvin C, Ninan I. Lack of experience-dependent intrinsic plasticity in the adolescent infralimbic medial prefrontal cortex. Synapse 2019; 73:e22090. [PMID: 30720888 DOI: 10.1002/syn.22090] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Revised: 01/12/2019] [Accepted: 02/01/2019] [Indexed: 01/24/2023]
Abstract
Fear extinction, an inhibitory learning that suppresses a previously learned fear memory, is diminished during adolescence. Earlier studies have shown that this suppressed fear extinction during adolescence involves an altered glutamatergic plasticity in infralimbic medial prefrontal cortical (IL-mPFC) pyramidal neurons. However, it is unclear whether the excitability of IL-mPFC pyramidal neurons plays a role in this development-dependent suppression of fear extinction. Therefore, we examined whether fear conditioning and extinction affect the active and passive membrane properties of IL-mPFC layer 5 pyramidal neurons in preadolescent, adolescent and adult mice. Both preadolescent and adult mice exhibited a bidirectional modulation of the excitability of IL-mPFC layer 5 pyramidal neurons following fear conditioning and extinction, i.e., fear conditioning reduced membrane excitability, whereas fear extinction reversed this effect. However, the fear conditioning-induced suppression of excitability was not reversed in adolescent mice following fear extinction training. Neither fear conditioning nor extinction affected GABAergic transmission in IL-mPFC layer 5 pyramidal neurons, suggesting that GABAergic transmission did not play a role in experience-dependent modulation of neuronal excitability. Our results suggest that the extinction-specific modulation of excitability is impaired during adolescence.
Collapse
Affiliation(s)
- Peter Koppensteiner
- Department of Psychiatry, NYU Neuroscience Institute, NYU Langone Medical Center, New York, New York
| | - Christopher Galvin
- Department of Psychiatry, NYU Neuroscience Institute, NYU Langone Medical Center, New York, New York
| | - Ipe Ninan
- Department of Psychiatry, NYU Neuroscience Institute, NYU Langone Medical Center, New York, New York
| |
Collapse
|
171
|
Deneault E, Faheem M, White SH, Rodrigues DC, Sun S, Wei W, Piekna A, Thompson T, Howe JL, Chalil L, Kwan V, Walker S, Pasceri P, Roth FP, Yuen RK, Singh KK, Ellis J, Scherer SW. CNTN5-/+or EHMT2-/+human iPSC-derived neurons from individuals with autism develop hyperactive neuronal networks. eLife 2019; 8:40092. [PMID: 30747104 PMCID: PMC6372285 DOI: 10.7554/elife.40092] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 01/19/2019] [Indexed: 12/14/2022] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived neurons are increasingly used to model Autism Spectrum Disorder (ASD), which is clinically and genetically heterogeneous. To study the complex relationship of penetrant and weaker polygenic risk variants to ASD, 'isogenic' iPSC-derived neurons are critical. We developed a set of procedures to control for heterogeneity in reprogramming and differentiation, and generated 53 different iPSC-derived glutamatergic neuronal lines from 25 participants from 12 unrelated families with ASD. Heterozygous de novo and rare-inherited presumed-damaging variants were characterized in ASD risk genes/loci. Combinations of putative etiologic variants (GLI3/KIF21A or EHMT2/UBE2I) in separate families were modeled. We used a multi-electrode array, with patch-clamp recordings, to determine a reproducible synaptic phenotype in 25% of the individuals with ASD (other relevant data on the remaining lines was collected). Our most compelling new results revealed a consistent spontaneous network hyperactivity in neurons deficient for CNTN5 or EHMT2. The biobank of iPSC-derived neurons and accompanying genomic data are available to accelerate ASD research. Editorial note This article has been through an editorial process in which authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Eric Deneault
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, Canada.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Muhammad Faheem
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, Canada.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Sean H White
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Canada
| | - Deivid C Rodrigues
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Song Sun
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada.,The Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Wei Wei
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Alina Piekna
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Tadeo Thompson
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Jennifer L Howe
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, Canada.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Leon Chalil
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Canada
| | - Vickie Kwan
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Canada
| | - Susan Walker
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, Canada.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Peter Pasceri
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, Canada
| | - Frederick P Roth
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Canada.,The Donnelly Centre, University of Toronto, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Department of Computer Science, University of Toronto, Toronto, Canada.,Canadian Institute for Advanced Research (CIFAR), Toronto, Canada
| | - Ryan Kc Yuen
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, Canada.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada
| | - Karun K Singh
- Department of Biochemistry and Biomedical Sciences, Stem Cell and Cancer Research Institute, McMaster University, Hamilton, Canada
| | - James Ellis
- Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Stephen W Scherer
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, Canada.,The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada.,McLaughlin Centre, University of Toronto, Toronto, Canada
| |
Collapse
|
172
|
Weykopf B, Haupt S, Jungverdorben J, Flitsch LJ, Hebisch M, Liu G, Suzuki K, Belmonte JCI, Peitz M, Blaess S, Till A, Brüstle O. Induced pluripotent stem cell-based modeling of mutant LRRK2-associated Parkinson's disease. Eur J Neurosci 2019; 49:561-589. [PMID: 30656775 PMCID: PMC7114274 DOI: 10.1111/ejn.14345] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 12/13/2018] [Accepted: 01/10/2019] [Indexed: 12/13/2022]
Abstract
Recent advances in cell reprogramming have enabled assessment of disease-related cellular traits in patient-derived somatic cells, thus providing a versatile platform for disease modeling and drug development. Given the limited access to vital human brain cells, this technology is especially relevant for neurodegenerative disorders such as Parkinson's disease (PD) as a tool to decipher underlying pathomechanisms. Importantly, recent progress in genome-editing technologies has provided an ability to analyze isogenic induced pluripotent stem cell (iPSC) pairs that differ only in a single genetic change, thus allowing a thorough assessment of the molecular and cellular phenotypes that result from monogenetic risk factors. In this review, we summarize the current state of iPSC-based modeling of PD with a focus on leucine-rich repeat kinase 2 (LRRK2), one of the most prominent monogenetic risk factors for PD linked to both familial and idiopathic forms. The LRRK2 protein is a primarily cytosolic multi-domain protein contributing to regulation of several pathways including autophagy, mitochondrial function, vesicle transport, nuclear architecture and cell morphology. We summarize iPSC-based studies that contributed to improving our understanding of the function of LRRK2 and its variants in the context of PD etiopathology. These data, along with results obtained in our own studies, underscore the multifaceted role of LRRK2 in regulating cellular homeostasis on several levels, including proteostasis, mitochondrial dynamics and regulation of the cytoskeleton. Finally, we expound advantages and limitations of reprogramming technologies for disease modeling and drug development and provide an outlook on future challenges and expectations offered by this exciting technology.
Collapse
Affiliation(s)
- Beatrice Weykopf
- Institute of Reconstructive NeurobiologyUniversity of Bonn School of Medicine & University Hospital BonnBonnGermany
- Life & Brain GmbHCellomics UnitBonnGermany
- Precision Neurology Program & Advanced Center for Parkinson's Disease ResearchHarvard Medical School and Brigham & Women's HospitalBostonMassachusetts
| | | | - Johannes Jungverdorben
- Institute of Reconstructive NeurobiologyUniversity of Bonn School of Medicine & University Hospital BonnBonnGermany
- Memorial Sloan Kettering Cancer CenterNew York CityNew York
| | - Lea Jessica Flitsch
- Institute of Reconstructive NeurobiologyUniversity of Bonn School of Medicine & University Hospital BonnBonnGermany
| | - Matthias Hebisch
- Institute of Reconstructive NeurobiologyUniversity of Bonn School of Medicine & University Hospital BonnBonnGermany
| | - Guang‐Hui Liu
- National Laboratory of BiomacromoleculesCAS Center for Excellence in BiomacromoleculesInstitute of BiophysicsChinese Academy of SciencesBeijingChina
| | - Keiichiro Suzuki
- Gene Expression LaboratoryThe Salk Institute for Biological StudiesLa JollaCalifornia
| | | | - Michael Peitz
- Institute of Reconstructive NeurobiologyUniversity of Bonn School of Medicine & University Hospital BonnBonnGermany
- German Center for Neurodegenerative Diseases (DZNE)BonnGermany
| | - Sandra Blaess
- Institute of Reconstructive NeurobiologyUniversity of Bonn School of Medicine & University Hospital BonnBonnGermany
| | - Andreas Till
- Institute of Reconstructive NeurobiologyUniversity of Bonn School of Medicine & University Hospital BonnBonnGermany
- Life & Brain GmbHCellomics UnitBonnGermany
| | - Oliver Brüstle
- Institute of Reconstructive NeurobiologyUniversity of Bonn School of Medicine & University Hospital BonnBonnGermany
| |
Collapse
|
173
|
Tanguay J, Callahan KM, D'Avanzo N. Characterization of drug binding within the HCN1 channel pore. Sci Rep 2019; 9:465. [PMID: 30679654 PMCID: PMC6345760 DOI: 10.1038/s41598-018-37116-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Accepted: 11/29/2018] [Indexed: 11/09/2022] Open
Abstract
Hyperpolarization-activated cyclic nucleotide-gated (HCN) channels mediate rhythmic electrical activity of cardiac pacemaker cells, and in neurons play important roles in setting resting membrane potentials, dendritic integration, neuronal pacemaking, and establishing action potential threshold. Block of HCN channels slows the heart rate and is currently used to treat angina. However, HCN block also provides a promising approach to the treatment of neuronal disorders including epilepsy and neuropathic pain. While several molecules that block HCN channels have been identified, including clonidine and its derivative alinidine, lidocaine, mepivacaine, bupivacaine, ZD7288, ivabradine, zatebradine, and cilobradine, their low affinity and lack of specificity prevents wide-spread use. Different studies suggest that the binding sites of these inhibitors are located in the inner vestibule of HCN channels, but the molecular details of their binding remain unknown. We used computational docking experiments to assess the binding sites and mode of binding of these inhibitors against the recently solved atomic structure of human HCN1 channels, and a homology model of the open pore derived from a closely related CNG channel. We identify a possible hydrophobic groove in the pore cavity that plays an important role in conformationally restricting the location and orientation of drugs bound to the inner vestibule. Our results also help explain the molecular basis of the low-affinity binding of these inhibitors, paving the way for the development of higher affinity molecules.
Collapse
Affiliation(s)
- Jérémie Tanguay
- Department of Physics, Université de Montréal, Montréal, Canada
| | - Karen M Callahan
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada
| | - Nazzareno D'Avanzo
- Department of Pharmacology and Physiology, Université de Montréal, Montréal, Canada.
| |
Collapse
|
174
|
Gouder L, Vitrac A, Goubran-Botros H, Danckaert A, Tinevez JY, André-Leroux G, Atanasova E, Lemière N, Biton A, Leblond CS, Poulet A, Boland A, Deleuze JF, Benchoua A, Delorme R, Bourgeron T, Cloëz-Tayarani I. Altered spinogenesis in iPSC-derived cortical neurons from patients with autism carrying de novo SHANK3 mutations. Sci Rep 2019; 9:94. [PMID: 30643170 PMCID: PMC6331634 DOI: 10.1038/s41598-018-36993-x] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 11/28/2018] [Indexed: 01/19/2023] Open
Abstract
The synaptic protein SHANK3 encodes a multidomain scaffold protein expressed at the postsynaptic density of neuronal excitatory synapses. We previously identified de novo SHANK3 mutations in patients with autism spectrum disorders (ASD) and showed that SHANK3 represents one of the major genes for ASD. Here, we analyzed the pyramidal cortical neurons derived from induced pluripotent stem cells from four patients with ASD carrying SHANK3 de novo truncating mutations. At 40-45 days after the differentiation of neural stem cells, dendritic spines from pyramidal neurons presented variable morphologies: filopodia, thin, stubby and muschroom, as measured in 3D using GFP labeling and immunofluorescence. As compared to three controls, we observed a significant decrease in SHANK3 mRNA levels (less than 50% of controls) in correlation with a significant reduction in dendritic spine densities and whole spine and spine head volumes. These results, obtained through the analysis of de novo SHANK3 mutations in the patients' genomic background, provide further support for the presence of synaptic abnormalities in a subset of patients with ASD.
Collapse
Affiliation(s)
- Laura Gouder
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France.,CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - Aline Vitrac
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France.,CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - Hany Goubran-Botros
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France.,CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | | | | | | | - Ekaterina Atanasova
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France.,CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - Nathalie Lemière
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France.,CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - Anne Biton
- Bioinformatics and Biostatistics Hub, C3BI, USR 3756 IP CNRS, Institut Pasteur, Paris, France
| | - Claire S Leblond
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France.,CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | | | - Anne Boland
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, F-91057, Evry, France
| | - Jean-François Deleuze
- Centre National de Recherche en Génomique Humaine (CNRGH), Institut de Biologie François Jacob, CEA, Université Paris-Saclay, F-91057, Evry, France
| | | | - Richard Delorme
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France.,Assistance Publique-Hôpitaux de Paris, Robert Debré Hospital, Department of Child and Adolescent Psychiatry, Paris, France
| | - Thomas Bourgeron
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France.,CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France
| | - Isabelle Cloëz-Tayarani
- Human Genetics and Cognitive Functions, Institut Pasteur, Paris, France. .,CNRS UMR 3571 « Genes, Synapses and Cognition », Institut Pasteur, Paris, France. .,Université Paris Diderot, Sorbonne Paris Cité, Human Genetics and Cognitive Functions, Paris, France.
| |
Collapse
|
175
|
Fantuzzo JA, Hart RP, Zahn JD, Pang ZP. Compartmentalized Devices as Tools for Investigation of Human Brain Network Dynamics. Dev Dyn 2019; 248:65-77. [PMID: 30117633 PMCID: PMC6312734 DOI: 10.1002/dvdy.24665] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Revised: 07/27/2018] [Accepted: 07/27/2018] [Indexed: 12/18/2022] Open
Abstract
Neuropsychiatric disorders have traditionally been difficult to study due to the complexity of the human brain and limited availability of human tissue. Induced pluripotent stem (iPS) cells provide a promising avenue to further our understanding of human disease mechanisms, but traditional 2D cell cultures can only provide a limited view of the neural circuits. To better model complex brain neurocircuitry, compartmentalized culturing systems and 3D organoids have been developed. Early compartmentalized devices demonstrated how neuronal cell bodies can be isolated both physically and chemically from neurites. Soft lithographic approaches have advanced this approach and offer the tools to construct novel model platforms, enabling circuit-level studies of disease, which can accelerate mechanistic studies and drug candidate screening. In this review, we describe some of the common technologies used to develop such systems and discuss how these lithographic techniques have been used to advance our understanding of neuropsychiatric disease. Finally, we address other in vitro model platforms such as 3D culture systems and organoids and compare these models with compartmentalized models. We ask important questions regarding how we can further harness iPS cells in these engineered culture systems for the development of improved in vitro models. Developmental Dynamics 248:65-77, 2019. © 2018 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Joseph A Fantuzzo
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, New Jersey
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Ronald P Hart
- Department of Cell Biology and Neuroscience, Rutgers University, Piscataway, New Jersey
| | - Jeffrey D Zahn
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey
| | - Zhiping P Pang
- Child Health Institute of New Jersey, Robert Wood Johnson Medical School, New Brunswick, New Jersey
- Department of Neuroscience and Cell Biology, Research Tower, Piscataway, New Jersey
- Pediatrics, Robert Wood Johnson Medical School, Rutgers University, New Brunswick, New Jersey
| |
Collapse
|
176
|
Kaffman A, White JD, Wei L, Johnson FK, Krystal JH. Enhancing the Utility of Preclinical Research in Neuropsychiatry Drug Development. Methods Mol Biol 2019; 2011:3-22. [PMID: 31273690 DOI: 10.1007/978-1-4939-9554-7_1] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Most large pharmaceutical companies have downscaled or closed their clinical neuroscience research programs in response to the low clinical success rate for drugs that showed tremendous promise in animal experiments intended to model psychiatric pathophysiology. These failures have raised serious concerns about the role of preclinical research in the identification and evaluation of new pharmacotherapies for psychiatry. In the absence of a comprehensive understanding of the neurobiology of psychiatric disorders, the task of developing "animal models" seems elusive. The purpose of this review is to highlight emerging strategies to enhance the utility of preclinical research in the drug development process. We address this issue by reviewing how advances in neuroscience, coupled with new conceptual approaches, have recently revolutionized the way we can diagnose and treat common psychiatric conditions. We discuss the implications of these new tools for modeling psychiatric conditions in animals and advocate for the use of systematic reviews of preclinical work as a prerequisite for conducting psychiatric clinical trials. We believe that work in animals is essential for elucidating human psychopathology and that improving the predictive validity of animal models is necessary for developing more effective interventions for mental illness.
Collapse
Affiliation(s)
- Arie Kaffman
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA.
| | - Jordon D White
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Lan Wei
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Frances K Johnson
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - John H Krystal
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
177
|
Du X, Gao X, Liu X, Shen L, Wang K, Fan Y, Sun Y, Luo X, Liu H, Wang L, Wang Y, Gong Z, Wang J, Yu Y, Li F. Genetic Diagnostic Evaluation of Trio-Based Whole Exome Sequencing Among Children With Diagnosed or Suspected Autism Spectrum Disorder. Front Genet 2018; 9:594. [PMID: 30555518 PMCID: PMC6284054 DOI: 10.3389/fgene.2018.00594] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 11/15/2018] [Indexed: 12/27/2022] Open
Abstract
Autism spectrum disorder (ASD) is a group of clinically and genetically heterogeneous neurodevelopmental disorders. Recent tremendous advances in the whole exome sequencing (WES) enable rapid identification of variants associated with ASD including single nucleotide variations (SNVs) and indels. To further explore genetic etiology of ASD in Chinese children with negative findings of copy number variants (CNVs), we applied WES in 80 simplex families with a single affected offspring with ASD or suspected ASD, and validated variations predicted to be damaging by Sanger sequencing. The results showed that an overall diagnostic yield of 8.8% (9.2% in the group of ASD and 6.7% in the group of suspected ASD) was observed in our cohort. Among patients with diagnosed ASD, developmental delay or intellectual disability (DD/ID) was the most common comorbidity with a diagnostic yield of 13.3%, followed by seizures (50.0%) and craniofacial anomalies (40.0%). All of identified de novo SNVs and indels among patients with ASD were loss of function (LOF) variations and were slightly more frequent among female (male vs. female: 7.3% vs. 8.5%). A total of seven presumed causative genes (CHD8, AFF2, ADNP, POGZ, SHANK3, IL1RAPL1, and PTEN) were identified in this study. In conclusion, WES is an efficient diagnostic tool for diagnosed ASD especially those with negative findings of CNVs and other neurological disorders in clinical practice, enabling early identification of disease related genes and contributing to precision and personalized medicine.
Collapse
Affiliation(s)
- Xiujuan Du
- Developmental and Behavioral Pediatric Department - Child Primary Care Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and MOE Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueren Gao
- Department of Pediatric Endocrinology and Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xin Liu
- Developmental and Behavioral Pediatric Department - Child Primary Care Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lixiao Shen
- Developmental and Behavioral Pediatric Department - Child Primary Care Department, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kai Wang
- Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yanjie Fan
- Department of Pediatric Endocrinology and Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Sun
- Department of Pediatric Endocrinology and Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaomei Luo
- Department of Pediatric Endocrinology and Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huili Liu
- Department of Pediatric Endocrinology and Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lili Wang
- Department of Pediatric Endocrinology and Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Wang
- Department of Pediatric Endocrinology and Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhuwen Gong
- Department of Pediatric Endocrinology and Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jianguo Wang
- Department of Pediatric Endocrinology and Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yongguo Yu
- Department of Pediatric Endocrinology and Genetics, Shanghai Institute for Pediatric Research, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Fei Li
- Developmental and Behavioral Pediatric Department - Child Primary Care Department, Brain and Behavioral Research Unit of Shanghai Institute for Pediatric Research and MOE Shanghai Key Laboratory for Children's Environmental Health, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
178
|
Srikanth P, Lagomarsino VN, Pearse RV, Liao M, Ghosh S, Nehme R, Seyfried N, Eggan K, Young-Pearse TL. Convergence of independent DISC1 mutations on impaired neurite growth via decreased UNC5D expression. Transl Psychiatry 2018; 8:245. [PMID: 30410030 PMCID: PMC6224395 DOI: 10.1038/s41398-018-0281-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/16/2018] [Indexed: 12/29/2022] Open
Abstract
The identification of convergent phenotypes in different models of psychiatric illness highlights robust phenotypes that are more likely to be implicated in disease pathophysiology. Here, we utilize human iPSCs harboring distinct mutations in DISC1 that have been found in families with major mental illness. One mutation was engineered to mimic the consequences on DISC1 protein of a balanced translocation linked to mental illness in a Scottish pedigree; the other mutation was identified in an American pedigree with a high incidence of mental illness. Directed differentiation of these iPSCs using NGN2 expression shows rapid conversion to a homogenous population of mature excitatory neurons. Both DISC1 mutations result in reduced DISC1 protein expression, and show subtle effects on certain presynaptic proteins. In addition, RNA sequencing and qPCR showed decreased expression of UNC5D, DPP10, PCDHA6, and ZNF506 in neurons with both DISC1 mutations. Longitudinal analysis of neurite outgrowth revealed decreased neurite outgrowth in neurons with each DISC1 mutation, which was mimicked by UNC5D knockdown and rescued by transient upregulation of endogenous UNC5D. This study shows a narrow range of convergent phenotypes of two mutations found in families with major mental illness, and implicates dysregulated netrin signaling in DISC1 biology.
Collapse
Affiliation(s)
- Priya Srikanth
- 0000 0004 0378 8294grid.62560.37Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Valentina N. Lagomarsino
- 0000 0004 0378 8294grid.62560.37Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Richard V. Pearse
- 0000 0004 0378 8294grid.62560.37Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Meichen Liao
- 0000 0004 0378 8294grid.62560.37Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| | - Sulagna Ghosh
- 000000041936754Xgrid.38142.3cHarvard Stem Cell Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA ,grid.66859.34Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Ralda Nehme
- 000000041936754Xgrid.38142.3cHarvard Stem Cell Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA ,grid.66859.34Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Nicholas Seyfried
- 0000 0001 0941 6502grid.189967.8Department of Biochemistry, Department of Neurology, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Kevin Eggan
- 000000041936754Xgrid.38142.3cHarvard Stem Cell Institute, Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138 USA ,grid.66859.34Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142 USA
| | - Tracy L. Young-Pearse
- 0000 0004 0378 8294grid.62560.37Ann Romney Center for Neurologic Diseases, Brigham and Women’s Hospital and Harvard Medical School, Boston, MA USA
| |
Collapse
|
179
|
Patel J, Lukkes JL, Shekhar A. Overview of genetic models of autism spectrum disorders. PROGRESS IN BRAIN RESEARCH 2018; 241:1-36. [PMID: 30447752 DOI: 10.1016/bs.pbr.2018.10.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Autism spectrum disorders (ASDs) are a group of neurodevelopment disorders that are characterized by heterogenous cognitive deficits and genetic factors. As more ASD risk genes are identified, genetic animal models have been developed to parse out the underlying neurobiological mechanisms of ASD. In this review, we discuss a subset of genetic models of ASD, focusing on those that have been widely studied and strongly linked to ASD. We focus our discussion of these models in the context of the theories and potential mechanisms of ASD, including disruptions in cell growth and proliferation, spine dynamics, synaptic transmission, excitation/inhibition balance, intracellular signaling, neuroinflammation, and behavior. In addition to ASD pathophysiology, we examine the limitations and challenges that genetic models pose for the study of ASD biology. We end with a review of innovative techniques and concepts of ASD pathology that can be further applied to and studied using genetic ASD models.
Collapse
Affiliation(s)
- Jheel Patel
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN, United States
| | - Jodi L Lukkes
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Anantha Shekhar
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States; Program in Medical Neuroscience, Paul and Carole Stark Neurosciences Research Institute, Indianapolis, IN, United States; Indiana Clinical and Translation Sciences Institute, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
180
|
Deneault E, White SH, Rodrigues DC, Ross PJ, Faheem M, Zaslavsky K, Wang Z, Alexandrova R, Pellecchia G, Wei W, Piekna A, Kaur G, Howe JL, Kwan V, Thiruvahindrapuram B, Walker S, Lionel AC, Pasceri P, Merico D, Yuen RKC, Singh KK, Ellis J, Scherer SW. Complete Disruption of Autism-Susceptibility Genes by Gene Editing Predominantly Reduces Functional Connectivity of Isogenic Human Neurons. Stem Cell Reports 2018; 11:1211-1225. [PMID: 30392976 PMCID: PMC6235011 DOI: 10.1016/j.stemcr.2018.10.003] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Revised: 10/01/2018] [Accepted: 10/02/2018] [Indexed: 12/31/2022] Open
Abstract
Autism spectrum disorder (ASD) is phenotypically and genetically heterogeneous. We present a CRISPR gene editing strategy to insert a protein tag and premature termination sites creating an induced pluripotent stem cell (iPSC) knockout resource for functional studies of ten ASD-relevant genes (AFF2/FMR2, ANOS1, ASTN2, ATRX, CACNA1C, CHD8, DLGAP2, KCNQ2, SCN2A, TENM1). Neurogenin 2 (NGN2)-directed induction of iPSCs allowed production of excitatory neurons, and mutant proteins were not detectable. RNA sequencing revealed convergence of several neuronal networks. Using both patch-clamp and multi-electrode array approaches, the electrophysiological deficits measured were distinct for different mutations. However, they culminated in a consistent reduction in synaptic activity, including reduced spontaneous excitatory postsynaptic current frequencies in AFF2/FMR2-, ASTN2-, ATRX-, KCNQ2-, and SCN2A-null neurons. Despite ASD susceptibility genes belonging to different gene ontologies, isogenic stem cell resources can reveal common functional phenotypes, such as reduced functional connectivity. iPSC knockout resource for functional studies of ten ASD-risk genes Disruption of common transcriptional networks associated with neurons and synapses Reduced synaptic activity commonly observed for functionally diverse ASD-risk genes
Collapse
Affiliation(s)
- Eric Deneault
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Sean H White
- Stem Cell and Cancer Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8S 4L8, Canada
| | - Deivid C Rodrigues
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - P Joel Ross
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Muhammad Faheem
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Kirill Zaslavsky
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3H7, Canada
| | - Zhuozhi Wang
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Roumiana Alexandrova
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Giovanna Pellecchia
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Wei Wei
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Alina Piekna
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Gaganjot Kaur
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Jennifer L Howe
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Vickie Kwan
- Stem Cell and Cancer Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8S 4L8, Canada
| | | | - Susan Walker
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Anath C Lionel
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Peter Pasceri
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Daniele Merico
- The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Ryan K C Yuen
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Karun K Singh
- Stem Cell and Cancer Research Institute, Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton L8S 4L8, Canada.
| | - James Ellis
- Developmental & Stem Cell Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3H7, Canada.
| | - Stephen W Scherer
- Genetics & Genome Biology Program, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; The Centre for Applied Genomics, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON M5S 3H7, Canada; McLaughlin Centre, University of Toronto, Toronto, ON M5S 3H7, Canada.
| |
Collapse
|
181
|
Shen X, Yeung HT, Lai KO. Application of Human-Induced Pluripotent Stem Cells (hiPSCs) to Study Synaptopathy of Neurodevelopmental Disorders. Dev Neurobiol 2018; 79:20-35. [PMID: 30304570 DOI: 10.1002/dneu.22644] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 09/27/2018] [Accepted: 10/04/2018] [Indexed: 12/15/2022]
Abstract
Synapses are the basic structural and functional units for information processing and storage in the brain. Their diverse properties and functions ultimately underlie the complexity of human behavior. Proper development and maintenance of synapses are essential for normal functioning of the nervous system. Disruption in synaptogenesis and the consequent alteration in synaptic function have been strongly implicated to cause neurodevelopmental disorders such as autism spectrum disorders (ASDs) and schizophrenia (SCZ). The introduction of human-induced pluripotent stem cells (hiPSCs) provides a new path to elucidate disease mechanisms and potential therapies. In this review, we will discuss the advantages and limitations of using hiPSC-derived neurons to study synaptic disorders. Many mutations in genes encoding for proteins that regulate synaptogenesis have been identified in patients with ASDs and SCZ. We use Methyl-CpG binding protein 2 (MECP2), SH3 and multiple ankyrin repeat domains 3 (SHANK3) and Disrupted in schizophrenia 1 (DISC1) as examples to illustrate the promise of using hiPSCs as cellular models to elucidate the mechanisms underlying disease-related synaptopathy.
Collapse
Affiliation(s)
- Xuting Shen
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Hoi Ting Yeung
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| | - Kwok-On Lai
- Faculty of Medicine, School of Biomedical Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China.,State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, 21 Sassoon Road, Hong Kong, China
| |
Collapse
|
182
|
Brumback AC, Elwood I, Kjaerby C, Iafrati J, Robinson S, Lee A, Patel T, Nagaraj S, Davatolhagh F, Sohal VS. Identifying specific prefrontal neurons that contribute to autism-associated abnormalities in physiology and social behavior. Mol Psychiatry 2018; 23:2078-2089. [PMID: 29112191 PMCID: PMC6594833 DOI: 10.1038/mp.2017.213] [Citation(s) in RCA: 111] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2017] [Accepted: 08/07/2017] [Indexed: 12/18/2022]
Abstract
Functional imaging and gene expression studies both implicate the medial prefrontal cortex (mPFC), particularly deep-layer projection neurons, as a potential locus for autism pathology. Here, we explored how specific deep-layer prefrontal neurons contribute to abnormal physiology and behavior in mouse models of autism. First, we find that across three etiologically distinct models-in utero valproic acid (VPA) exposure, CNTNAP2 knockout and FMR1 knockout-layer 5 subcortically projecting (SC) neurons consistently exhibit reduced input resistance and action potential firing. To explore how altered SC neuron physiology might impact behavior, we took advantage of the fact that in deep layers of the mPFC, dopamine D2 receptors (D2Rs) are mainly expressed by SC neurons, and used D2-Cre mice to label D2R+ neurons for calcium imaging or optogenetics. We found that social exploration preferentially recruits mPFC D2R+ cells, but that this recruitment is attenuated in VPA-exposed mice. Stimulating mPFC D2R+ neurons disrupts normal social interaction. Conversely, inhibiting these cells enhances social behavior in VPA-exposed mice. Importantly, this effect was not reproduced by nonspecifically inhibiting mPFC neurons in VPA-exposed mice, or by inhibiting D2R+ neurons in wild-type mice. These findings suggest that multiple forms of autism may alter the physiology of specific deep-layer prefrontal neurons that project to subcortical targets. Furthermore, a highly overlapping population-prefrontal D2R+ neurons-plays an important role in both normal and abnormal social behavior, such that targeting these cells can elicit potentially therapeutic effects.
Collapse
Affiliation(s)
- Audrey C. Brumback
- Department of Neurology, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.,Department of Psychiatry, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA.,Current address: Departments of Neurology and Pediatrics, Dell Medical School, and Center for Learning and Memory at The University of Texas at Austin
| | - Ian Elwood
- Department of Psychiatry, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Celia Kjaerby
- Department of Psychiatry, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Jillian Iafrati
- Department of Psychiatry, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Sarah Robinson
- Department of Psychiatry, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Anthony Lee
- Department of Psychiatry, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Tosha Patel
- Department of Psychiatry, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Suraj Nagaraj
- Department of Psychiatry, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Felicia Davatolhagh
- Department of Psychiatry, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| | - Vikaas S. Sohal
- Department of Psychiatry, Weill Institute for Neurosciences, Kavli Institute for Fundamental Neuroscience, University of California, San Francisco, San Francisco, CA, USA
| |
Collapse
|
183
|
Zatkova M, Bacova Z, Puerta F, Lestanova Z, Alanazi M, Kiss A, Reichova A, Castejon AM, Ostatnikova D, Bakos J. Projection length stimulated by oxytocin is modulated by the inhibition of calcium signaling in U-87MG cells. J Neural Transm (Vienna) 2018; 125:1847-1856. [DOI: 10.1007/s00702-018-1933-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 09/21/2018] [Indexed: 12/12/2022]
|
184
|
Fisher DW, Han Y, Lyman KA, Heuermann RJ, Bean LA, Ybarra N, Foote KM, Dong H, Nicholson DA, Chetkovich DM. HCN channels in the hippocampus regulate active coping behavior. J Neurochem 2018; 146:753-766. [PMID: 29953635 PMCID: PMC6158061 DOI: 10.1111/jnc.14539] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Revised: 06/05/2018] [Accepted: 06/07/2018] [Indexed: 12/16/2022]
Abstract
Active coping is an adaptive stress response that improves outcomes in medical and neuropsychiatric diseases. To date, most research into coping style has focused on neurotransmitter activity and little is known about the intrinsic excitability of neurons in the associated brain regions that facilitate coping. Previous studies have shown that HCN channels regulate neuronal excitability in pyramidal cells and that HCN channel current (Ih ) in the CA1 area increases with chronic mild stress. Reduction of Ih in the CA1 area leads to antidepressant-like behavior, and this region has been implicated in the regulation of coping style. We hypothesized that the antidepressant-like behavior achieved with CA1 knockdown of Ih is accompanied by increases in active coping. In this report, we found that global loss of TRIP8b, a necessary subunit for proper HCN channel localization in pyramidal cells, led to active coping behavior in numerous assays specific to coping style. We next employed a viral strategy using a dominant negative TRIP8b isoform to alter coping behavior by reducing HCN channel expression. This approach led to a robust reduction in Ih in CA1 pyramidal neurons and an increase in active coping. Together, these results establish that changes in HCN channel function in CA1 influences coping style.
Collapse
Affiliation(s)
- Daniel W Fisher
- Davee Department of Neurology and Clinical Neurosciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Ye Han
- Davee Department of Neurology and Clinical Neurosciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Kyle A Lyman
- Davee Department of Neurology and Clinical Neurosciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Robert J Heuermann
- Davee Department of Neurology and Clinical Neurosciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Linda A Bean
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Natividad Ybarra
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Kendall M Foote
- Davee Department of Neurology and Clinical Neurosciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
| | - Hongxin Dong
- Department of Psychiatry & Behavioral Sciences, Northwestern University, Feinberg School of medicine, Chicago, Illinois, USA
| | - Daniel A Nicholson
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois, USA
| | - Dane M Chetkovich
- Davee Department of Neurology and Clinical Neurosciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, USA
- Department of Neurology, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| |
Collapse
|
185
|
Epigenetic and Cellular Diversity in the Brain through Allele-Specific Effects. Trends Neurosci 2018; 41:925-937. [PMID: 30098802 DOI: 10.1016/j.tins.2018.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Revised: 06/29/2018] [Accepted: 07/10/2018] [Indexed: 01/18/2023]
Abstract
The benefits of diploidy are considered to involve masking partially recessive mutations and increasing genetic diversity. Here, we review new studies showing evidence for diverse allele-specific expression and epigenetic states in mammalian brain cells, which suggest that diploidy expands the landscape of gene regulatory and expression programs in cells. Allele-specific expression has been thought to be restricted to a few specific classes of genes. However, new studies show novel genomic imprinting effects that are brain-region-, cell-type- and age-dependent. In addition, novel forms of random monoallelic expression that impact many autosomal genes have been described in vitro and in vivo. We discuss the implications for understanding the benefits of diploidy, and the mechanisms shaping brain development, function, and disease.
Collapse
|
186
|
Hassani Nia F, Kreienkamp HJ. Functional Relevance of Missense Mutations Affecting the N-Terminal Part of Shank3 Found in Autistic Patients. Front Mol Neurosci 2018; 11:268. [PMID: 30131675 PMCID: PMC6090658 DOI: 10.3389/fnmol.2018.00268] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/16/2018] [Indexed: 12/22/2022] Open
Abstract
Genetic defects in SHANK genes are associated with autism. Deletions and truncating mutations suggest haploinsufficiency for Shank3 as a major cause of disease which may be analyzed in appropriate Shank deficient mouse models. Here we will focus on the functional analysis of missense mutations found in SHANK genes. The relevance of most of these mutations for Shank function, and their role in autism pathogenesis is unclear. This is partly due to the fact that mutations spare the most well studied functional domains of Shank3, such as the PDZ and SAM domains, or the short proline-rich motifs which are required for interactions with postsynaptic partners Homer, Cortactin, dynamin, IRSp53 and Abi-1. One set of mutations affects the N-terminal part, including the highly conserved SPN domain and ankyrin repeats. Functional analysis from several groups has indicated that these mutations (e.g., R12C; L68P; R300C, and Q321R) interfere with the critical role of Shank3 for synapse formation. More recently the structural analysis of the SPN-ARR module has begun to shed light on the molecular consequences of mutations in the SPN of Shank3. The SPN was identified as a Ras association domain, with high affinities for GTP-bound, active forms of Ras and Rap. The two autism related mutations in this part of the protein, R12C and L68P, both abolish Ras binding. Further work is directed at identifying the consequences of Ras binding to Shank proteins at postsynaptic sites.
Collapse
Affiliation(s)
- Fatemeh Hassani Nia
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hans-Jürgen Kreienkamp
- Institute for Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
187
|
Lauber E, Filice F, Schwaller B. Dysregulation of Parvalbumin Expression in the Cntnap2-/- Mouse Model of Autism Spectrum Disorder. Front Mol Neurosci 2018; 11:262. [PMID: 30116174 PMCID: PMC6082962 DOI: 10.3389/fnmol.2018.00262] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2018] [Accepted: 07/12/2018] [Indexed: 01/06/2023] Open
Abstract
Due to the complex and heterogeneous etiology of autism spectrum disorder (ASD), identification of convergent pathways and/or common molecular endpoints in the pathophysiological processes of ASD development are highly needed in order to facilitate treatment approaches targeted at the core symptoms. We recently reported on decreased expression of the Ca2+-binding protein parvalbumin (PV) in three well-characterized ASD mouse models, Shank1-/-, Shank3B-/- and in utero VPA-exposed mice. Moreover, PV-deficient mice (PV+/- and PV-/-) were found to show behavioral impairments and neuroanatomical changes closely resembling those frequently found in human ASD individuals. Here, we combined a stereology-based approach with molecular biology methods to assess changes in the subpopulation of PV-expressing (Pvalb) interneurons in the recently characterized contactin-associated protein-like 2 (Cntnap2-/-) knockout mouse model of ASD. The CNTNAP2 gene codes for a synaptic cell adhesion molecule involved in neurodevelopmental processes; mutations affecting the human CNTNAP2 locus are associated with human ASD core symptoms, in particular speech and language problems. We demonstrate that in Cntnap2-/- mice, no loss of Pvalb neurons is evident in ASD-associated brain regions including the striatum, somatosensory cortex (SSC) and medial prefrontal cortex (mPFC), shown by the unaltered number of Pvalb neurons ensheathed by VVA-positive perineuronal nets. However, the number of PV-immunoreactive (PV+) neurons and also PV protein levels were decreased in the striatum of Cntnap2-/- mice indicating that PV expression levels in some striatal Pvalb neurons dropped below the detection limit, yet without a loss of Pvalb neurons. No changes in PV+ neuron numbers were detected in the cortical regions investigated and also cortical PV expression levels were unaltered. Considering that Cntnap2 shows high expression levels in the striatum during human and mouse embryonic development and that the cortico-striato-thalamic circuitry is important for speech and language development, alterations in striatal PV expression and associated (homeostatic) adaptations are likely to play an important role in Cntnap2-/- mice and, assumingly, in human ASD patients with known Cntnap2 mutations.
Collapse
Affiliation(s)
- Emanuel Lauber
- Anatomy Unit, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Federica Filice
- Anatomy Unit, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| | - Beat Schwaller
- Anatomy Unit, Section of Medicine, University of Fribourg, Fribourg, Switzerland
| |
Collapse
|
188
|
Daghsni M, Rima M, Fajloun Z, Ronjat M, Brusés JL, M'rad R, De Waard M. Autism throughout genetics: Perusal of the implication of ion channels. Brain Behav 2018; 8:e00978. [PMID: 29934975 PMCID: PMC6085908 DOI: 10.1002/brb3.978] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Revised: 03/01/2018] [Accepted: 03/18/2018] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) comprises a group of neurodevelopmental psychiatric disorders characterized by deficits in social interactions, interpersonal communication, repetitive and stereotyped behaviors and may be associated with intellectual disabilities. The description of ASD as a synaptopathology highlights the importance of the synapse and the implication of ion channels in the etiology of these disorders. METHODS A narrative and critical review of the relevant papers from 1982 to 2017 known by the authors was conducted. RESULTS Genome-wide linkages, association studies, and genetic analyses of patients with ASD have led to the identification of several candidate genes and mutations linked to ASD. Many of the candidate genes encode for proteins involved in neuronal development and regulation of synaptic function including ion channels and actors implicated in synapse formation. The involvement of ion channels in ASD is of great interest as they represent attractive therapeutic targets. In agreement with this view, recent findings have shown that drugs modulating ion channel function are effective for the treatment of certain types of patients with ASD. CONCLUSION This review describes the genetic aspects of ASD with a focus on genes encoding ion channels and highlights the therapeutic implications of ion channels in the treatment of ASD.
Collapse
Affiliation(s)
- Marwa Daghsni
- L'institut du Thorax, INSERM UMR1087/CNRS UMR6291, Université de Nantes, Nantes, France.,Université de Tunis El Manar, Faculté de Médecine de Tunis, LR99ES10 Laboratoire de Génétique Humaine, 1007, Tunis, Tunisie
| | - Mohamad Rima
- Department of Neuroscience, Institute of Biology Paris-Seine, CNRS UMR 8246, INSERM U1130, Sorbonne Universités, Paris, France
| | - Ziad Fajloun
- Azm Center for Research in Biotechnology and Its Application, Lebanese University, Tripoli, Lebanon
| | - Michel Ronjat
- L'institut du Thorax, INSERM UMR1087/CNRS UMR6291, Université de Nantes, Nantes, France.,LabEx Ion Channels Science and Therapeutics, Nice, France
| | - Juan L Brusés
- Department of Natural Sciences, Mercy College, Dobbs Ferry, NY, USA
| | - Ridha M'rad
- Université de Tunis El Manar, Faculté de Médecine de Tunis, LR99ES10 Laboratoire de Génétique Humaine, 1007, Tunis, Tunisie.,Service des Maladies Congénitales et Héréditaires, Hôpital Charles Nicolle, Tunis, Tunisie
| | - Michel De Waard
- L'institut du Thorax, INSERM UMR1087/CNRS UMR6291, Université de Nantes, Nantes, France.,LabEx Ion Channels Science and Therapeutics, Nice, France
| |
Collapse
|
189
|
Taylor SE, Taylor RD, Price J, Andreae LC. Single-molecule fluorescence in-situ hybridization reveals that human SHANK3 mRNA expression varies during development and in autism-associated SHANK3 heterozygosity. Stem Cell Res Ther 2018; 9:206. [PMID: 30064494 PMCID: PMC6069870 DOI: 10.1186/s13287-018-0957-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Revised: 06/13/2018] [Accepted: 07/16/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Deletions and mutations in the SHANK3 gene are strongly associated with autism spectrum disorder and underlie the autism-associated disorder Phelan-McDermid syndrome. SHANK3 is a scaffolding protein found at the post-synaptic membrane of excitatory neurons. METHODS Single-molecule fluorescence in-situ hybridization (smFISH) allows the visualization of single mRNA transcripts in vitro. Here we perform and quantify smFISH in human inducible pluripotent stem cell (hiPSC)-derived cortical neurons, targeting the SHANK3 transcript. RESULTS Both smFISH and conventional immunofluorescence staining demonstrated a developmental increase in SHANK3 mRNA and protein, respectively, in control human cortical neurons. Analysis of single SHANK3 mRNA molecules in neurons derived from an autistic individual heterozygous for SHANK3 indicated that while the number of SHANK3 mRNA transcripts remained comparable with control levels in the cell soma, there was a 50% reduction within neuronal processes, suggesting that local, dendritic targeting of SHANK3 mRNA may be specifically affected in SHANK3 haploinsufficiency. CONCLUSION Human SHANK3 mRNA shows developmentally regulated dendritic localization in hiPSC-derived neurons, which is reduced in neurons generated from a haploinsufficient individual with autism. Although further replication is needed, given the importance of local mRNA translation in synaptic function, this could represent an important early abnormality.
Collapse
Affiliation(s)
- Samuel E Taylor
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Ruth D Taylor
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK.,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Jack Price
- MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.,Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE5 8AF, UK
| | - Laura C Andreae
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, SE1 1UL, UK. .,MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
190
|
Roessler R, Goldmann J, Shivalila C, Jaenisch R. JIP2 haploinsufficiency contributes to neurodevelopmental abnormalities in human pluripotent stem cell-derived neural progenitors and cortical neurons. Life Sci Alliance 2018; 1:e201800094. [PMID: 30456368 PMCID: PMC6238622 DOI: 10.26508/lsa.201800094] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 06/06/2018] [Accepted: 06/07/2018] [Indexed: 11/24/2022] Open
Abstract
Molecular and cellular profiling of patient-specific neural cell types provides suggestions for the involvement of JIP2 in the neurodevelopmental disorder Phelan–McDermid syndrome. Phelan–McDermid syndrome (also known as 22q13.3 deletion syndrome) is a syndromic form of autism spectrum disorder and currently thought to be caused by heterozygous loss of SHANK3. However, patients most frequently present with large chromosomal deletions affecting several additional genes. We used human pluripotent stem cell technology and genome editing to further dissect molecular and cellular mechanisms. We found that loss of JIP2 (MAPK8IP2) may contribute to a distinct neurodevelopmental phenotype in neural progenitor cells (NPCs) affecting neuronal maturation. This is most likely due to a simultaneous down-regulation of c-Jun N-terminal kinase (JNK) proteins, leading to impaired generation of mature neurons. Furthermore, semaphorin signaling appears to be impaired in patient NPCs and neurons. Pharmacological activation of neuropilin receptor 1 (NRP1) rescued impaired semaphorin pathway activity and JNK expression in patient neurons. Our results suggest a novel disease-specific mechanism involving the JIP/JNK complex and identify NRP1 as a potential new therapeutic target.
Collapse
Affiliation(s)
| | | | | | - Rudolf Jaenisch
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA.,Department of Biology, Massachusetts Institute of Technology, Cambridge, MA, USA
| |
Collapse
|
191
|
Store depletion-induced h-channel plasticity rescues a channelopathy linked to Alzheimer's disease. Neurobiol Learn Mem 2018; 154:141-157. [PMID: 29906573 DOI: 10.1016/j.nlm.2018.06.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Revised: 05/25/2018] [Accepted: 06/11/2018] [Indexed: 12/20/2022]
Abstract
Voltage-gated ion channels are critical for neuronal integration. Some of these channels, however, are misregulated in several neurological disorders, causing both gain- and loss-of-function channelopathies in neurons. Using several transgenic mouse models of Alzheimer's disease (AD), we find that sub-threshold voltage signals strongly influenced by hyperpolarization-activated, cyclic nucleotide-gated (HCN) channels progressively deteriorate over chronological aging in hippocampal CA1 pyramidal neurons. The degraded signaling via HCN channels in the transgenic mice is accompanied by an age-related global loss of their non-uniform dendritic expression. Both the aberrant signaling via HCN channels and their mislocalization could be restored using a variety of pharmacological agents that target the endoplasmic reticulum (ER). Our rescue of the HCN channelopathy helps provide molecular details into the favorable outcomes of ER-targeting drugs on the pathogenesis and synaptic/cognitive deficits in AD mouse models, and implies that they might have beneficial effects on neurological disorders linked to HCN channelopathies.
Collapse
|
192
|
Wang X, Kery R, Xiong Q. Synaptopathology in autism spectrum disorders: Complex effects of synaptic genes on neural circuits. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:398-415. [PMID: 28986278 DOI: 10.1016/j.pnpbp.2017.09.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Revised: 09/05/2017] [Accepted: 09/26/2017] [Indexed: 01/03/2023]
Affiliation(s)
- Xinxing Wang
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY 11794, USA
| | - Rachel Kery
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY 11794, USA; Medical Scientist Training Program (MSTP), Stony Brook University, Stony Brook, NY 11794, USA
| | - Qiaojie Xiong
- Department of Neurobiology & Behavior, Stony Brook University, Stony Brook, NY 11794, USA.
| |
Collapse
|
193
|
Mossa A, Giona F, Pagano J, Sala C, Verpelli C. SHANK genes in autism: Defining therapeutic targets. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:416-423. [PMID: 29175319 DOI: 10.1016/j.pnpbp.2017.11.019] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Revised: 11/14/2017] [Accepted: 11/18/2017] [Indexed: 01/16/2023]
Affiliation(s)
- Adele Mossa
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Federica Giona
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Jessica Pagano
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Carlo Sala
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy
| | - Chiara Verpelli
- CNR Neuroscience Institute, Department of Biotechnology and Translational Medicine, University of Milan, Milan, Italy.
| |
Collapse
|
194
|
Linda K, Fiuza C, Nadif Kasri N. The promise of induced pluripotent stem cells for neurodevelopmental disorders. Prog Neuropsychopharmacol Biol Psychiatry 2018; 84:382-391. [PMID: 29128445 DOI: 10.1016/j.pnpbp.2017.11.009] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Revised: 10/30/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
A major challenge in clinical genetics and medicine is represented by genetically and phenotypically highly diverse neurodevelopmental disorders, like for example intellectual disability and autism. Intellectual disability is characterized by substantial limitations in cognitive function and adaptive behaviour. At the cellular level, this is reflected by deficits in synaptic structure and plasticity and therefore has been coined as a synaptic disorder or "synaptopathy". In this review, we summarize the findings from recent studies in which iPSCs have been used to model specific neurodevelopmental syndromes, including Fragile X syndrome, Rett syndrome, Williams-Beuren syndrome and Phelan-McDermid syndrome. We discuss what we have learned from these studies and what key issues need to be addressed to move the field forward.
Collapse
Affiliation(s)
- Katrin Linda
- Department of Human Genetics, Department of Cognitive Neuroscience, Radboudumc, 6500 HB, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Carol Fiuza
- Department of Human Genetics, Department of Cognitive Neuroscience, Radboudumc, 6500 HB, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Department of Cognitive Neuroscience, Radboudumc, 6500 HB, Nijmegen, The Netherlands; Donders Institute for Brain, Cognition, and Behaviour, Centre for Neuroscience, 6525 AJ, Nijmegen, The Netherlands.
| |
Collapse
|
195
|
Hazelbaker DZ, Beccard A, Bara AM, Dabkowski N, Messana A, Mazzucato P, Lam D, Manning D, Eggan K, Barrett LE. A Scaled Framework for CRISPR Editing of Human Pluripotent Stem Cells to Study Psychiatric Disease. Stem Cell Reports 2018; 9:1315-1327. [PMID: 29020615 PMCID: PMC5639480 DOI: 10.1016/j.stemcr.2017.09.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Revised: 09/08/2017] [Accepted: 09/11/2017] [Indexed: 12/22/2022] Open
Abstract
Scaling of CRISPR-Cas9 technology in human pluripotent stem cells (hPSCs) represents an important step for modeling complex disease and developing drug screens in human cells. However, variables affecting the scaling efficiency of gene editing in hPSCs remain poorly understood. Here, we report a standardized CRISPR-Cas9 approach, with robust benchmarking at each step, to successfully target and genotype a set of psychiatric disease-implicated genes in hPSCs and provide a resource of edited hPSC lines for six of these genes. We found that transcriptional state and nucleosome positioning around targeted loci was not correlated with editing efficiency. However, editing frequencies varied between different hPSC lines and correlated with genomic stability, underscoring the need for careful cell line selection and unbiased assessments of genomic integrity. Together, our step-by-step quantification and in-depth analyses provide an experimental roadmap for scaling Cas9-mediated editing in hPSCs to study psychiatric disease, with broader applicability for other polygenic diseases.
Collapse
Affiliation(s)
- Dane Z Hazelbaker
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Amanda Beccard
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Anne M Bara
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Nicole Dabkowski
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Angelica Messana
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Patrizia Mazzucato
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Daisy Lam
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Danielle Manning
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Kevin Eggan
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| | - Lindy E Barrett
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
196
|
Yakoub AM, Sadek M. Development and Characterization of Human Cerebral Organoids: An Optimized Protocol. Cell Transplant 2018; 27:393-406. [PMID: 29749250 PMCID: PMC6038047 DOI: 10.1177/0963689717752946] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Studies of human neurodevelopmental disorders and stem cell–based regenerative transplants have been hampered by the lack of a model of the developing human brain. Stem cell–derived neurons suffer major limitations, including the ability to recapitulate the 3-dimensional architecture of a brain tissue and the representation of multiple layers and cell types that contribute to the overall brain functions in vivo. Recently, cerebral organoid technology was introduced; however, such technology is still in its infancy, and its low reproducibility and limitations significantly reduce the reliability of such a model as it currently exists, especially considering the complexity of cerebral-organoid protocols. Here we have tested and compared multiple protocols and conditions for growth of organoids, and we describe an optimized methodology, and define the necessary and sufficient factors that support the development of optimal organoids. Our optimization criteria included organoids’ overall growth and size, stratification and representation of the various cell types, inter-batch variability, analysis of neuronal maturation, and even the cost of the procedure. Importantly, this protocol encompasses a plethora of technical tips that allow researchers to easily reproduce it and obtain reliable organoids with the least variability, and showcases a robust array of approaches to characterize successful organoids. This optimized protocol provides a reliable system for genetic or pharmacological (drug development) screens and may enhance understanding and therapy of human neurodevelopmental disorders, including harnessing the therapeutic potential of stem cell–derived transplants.
Collapse
Affiliation(s)
- Abraam M Yakoub
- 1 Department of Physiology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA
| | - Mark Sadek
- 2 Department of Pharmaceutical Biotechnology, University of Illinois College of Pharmacy, Chicago, IL, USA.,3 Department of Research and Development, Akorn Pharmaceuticals, Vernon Hills, IL, USA
| |
Collapse
|
197
|
Yang SS, Li YC, Coley AA, Chamberlin LA, Yu P, Gao WJ. Cell-Type Specific Development of the Hyperpolarization-Activated Current, Ih, in Prefrontal Cortical Neurons. Front Synaptic Neurosci 2018; 10:7. [PMID: 29867437 PMCID: PMC5958189 DOI: 10.3389/fnsyn.2018.00007] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 04/24/2018] [Indexed: 11/13/2022] Open
Abstract
H-current, also known as hyperpolarization-activated current (Ih), is an inward current generated by the hyperpolarization-activated cyclic nucleotide-gated (HCN) cation channels. Ih plays an essential role in regulating neuronal properties, synaptic integration and plasticity, and synchronous activity in the brain. As these biological factors change across development, the brain undergoes varying levels of vulnerability to disorders like schizophrenia that disrupt prefrontal cortex (PFC)-dependent function. However, developmental changes in Ih in PFC neurons remains untested. Here, we examine Ih in pyramidal neurons vs. gamma-aminobutyric acid (GABA)ergic parvalbumin-expressing (PV+) interneurons in developing mouse PFC. Our findings show that the amplitudes of Ih in these cell types are identical during the juvenile period but differ at later time points. In pyramidal neurons, Ih amplitude significantly increases from juvenile to adolescence and follows a similar trend into adulthood. In contrast, the amplitude of Ih in PV+ interneurons decreases from juvenile to adolescence, and does not change from adolescence to adulthood. Moreover, the kinetics of HCN channels in pyramidal neurons is significantly slower than in PV+ interneurons, with a gradual decrease in pyramidal neurons and a gradual increase in PV+ cells across development. Our study reveals distinct developmental trajectories of Ih in pyramidal neurons and PV+ interneurons. The cell-type specific alteration of Ih during the critical period from juvenile to adolescence reflects the contribution of Ih to the maturation of the PFC and PFC-dependent function. These findings are essential for a better understanding of normal PFC function, and for elucidating Ih's crucial role in the pathophysiology of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Sha-Sha Yang
- Beijing Key Laboratory of Learning and Cognition, College of Psychology, Capital Normal University, Beijing, China.,Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Yan-Chun Li
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Austin A Coley
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Linda A Chamberlin
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
| | - Ping Yu
- Beijing Key Laboratory of Learning and Cognition, College of Psychology, Capital Normal University, Beijing, China
| | - Wen-Jun Gao
- Department of Neurobiology and Anatomy, College of Medicine, Drexel University, Philadelphia, PA, United States
| |
Collapse
|
198
|
Kawasaki Y, Saito M, Won J, Bae JY, Sato H, Toyoda H, Kuramoto E, Kogo M, Tanaka T, Kaneko T, Oh SB, Bae YC, Kang Y. Inhibition of GluR Current in Microvilli of Sensory Neurons via Na +-Microdomain Coupling Among GluR, HCN Channel, and Na +/K + Pump. Front Cell Neurosci 2018; 12:113. [PMID: 29740287 PMCID: PMC5928758 DOI: 10.3389/fncel.2018.00113] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 04/06/2018] [Indexed: 11/13/2022] Open
Abstract
Glutamatergic dendritic EPSPs evoked in cortical pyramidal neurons are depressed by activation of hyperpolarization-activated cyclic nucleotide-gated (HCN) channels expressed in dendritic spines. This depression has been attributed to shunting effects of HCN current (Ih) on input resistance or Ih deactivation. Primary sensory neurons in the rat mesencephalic trigeminal nucleus (MTN) have the somata covered by spine-like microvilli that express HCN channels. In rat MTN neurons, we demonstrated that Ih enhancement apparently diminished the glutamate receptor (GluR) current (IGluR) evoked by puff application of glutamate/AMPA and enhanced a transient outward current following IGluR (OT-IGluR). This suggests that some outward current opposes inward IGluR. The IGluR inhibition displayed a U-shaped voltage-dependence with a minimal inhibition around the resting membrane potential, suggesting that simple shunting effects or deactivation of Ih cannot explain the U-shaped voltage-dependence. Confocal imaging of Na+ revealed that GluR activation caused an accumulation of Na+ in the microvilli, which can cause a negative shift of the reversal potential for Ih (Eh). Taken together, it was suggested that IGluR evoked in MTN neurons is opposed by a transient decrease or increase in standing inward or outward Ih, respectively, both of which can be caused by negative shifts of Eh, as consistent with the U-shaped voltage-dependence of the IGluR inhibition and the OT-IGluR generation. An electron-microscopic immunohistochemical study revealed the colocalization of HCN channels and glutamatergic synapses in microvilli of MTN neurons, which would provide a morphological basis for the functional interaction between HCN and GluR channels. Mathematical modeling eliminated the possibilities of the involvements of Ih deactivation and/or shunting effect and supported the negative shift of Eh which causes the U-shaped voltage-dependent inhibition of IGluR.
Collapse
Affiliation(s)
- Yasuhiro Kawasaki
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Mitsuru Saito
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Jonghwa Won
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Jin Young Bae
- Department of Oral Anatomy, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Hajime Sato
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Hiroki Toyoda
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Eriko Kuramoto
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Mikihiko Kogo
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| | - Takuma Tanaka
- Department of Computational Intelligence and Systems Science, Interdisciplinary Graduate School of Science and Engineering, Tokyo Institute of Technology, Yokohama, Japan
| | - Takeshi Kaneko
- Department of Morphological Brain Science, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Seog Bae Oh
- Department of Brain and Cognitive Sciences, College of Natural Sciences, Dental Research Institute and Department of Neurobiology and Physiology, School of Dentistry, Seoul National University, Seoul, South Korea
| | - Yong Chul Bae
- Department of Oral Anatomy, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Youngnam Kang
- Department of Neuroscience and Oral Physiology, Graduate School of Dentistry, Osaka University, Osaka, Japan
| |
Collapse
|
199
|
Abstract
Understanding the development of the human brain in relation with evolution is an important frontier field in developmental biology. In particular, investigating the mechanisms underlying the greatly increased relative size and complexity of the cerebral cortex, the seat of our enhanced cognitive abilities, remains a fascinating yet largely unsolved question. Though many advances in our understanding have been gained from the study of animal models, as well as human genetics and embryology, large gaps remain in our knowledge of the molecular mechanisms that control human cortical development. Interestingly, many aspects of corticogenesis can be recapitulated in vitro from mouse and human embryonic or induced pluripotent stem cells (PSCs), using a variety of experimental systems from 2D models to organoids to xenotransplantation. This has provided the opportunity to study these processes in an accessible and physiologically relevant setting. In this chapter, we will discuss how conserved and divergent features of primate/human corticogenesis can be modeled and studied mechanistically using PSC-based models of corticogenesis. We will also review what has been learned through these approaches about pathological defects of human corticogenesis, from early neurogenesis to late neuronal maturation and connectivity.
Collapse
|
200
|
Antunes G, Faria da Silva SF, Simoes de Souza FM. Mirror Neurons Modeled Through Spike-Timing-Dependent Plasticity are Affected by Channelopathies Associated with Autism Spectrum Disorder. Int J Neural Syst 2018; 28:1750058. [DOI: 10.1142/s0129065717500587] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Mirror neurons fire action potentials both when the agent performs a certain behavior and watches someone performing a similar action. Here, we present an original mirror neuron model based on the spike-timing-dependent plasticity (STDP) between two morpho-electrical models of neocortical pyramidal neurons. Both neurons fired spontaneously with basal firing rate that follows a Poisson distribution, and the STDP between them was modeled by the triplet algorithm. Our simulation results demonstrated that STDP is sufficient for the rise of mirror neuron function between the pairs of neocortical neurons. This is a proof of concept that pairs of neocortical neurons associating sensory inputs to motor outputs could operate like mirror neurons. In addition, we used the mirror neuron model to investigate whether channelopathies associated with autism spectrum disorder could impair the modeled mirror function. Our simulation results showed that impaired hyperpolarization-activated cationic currents (Ih) affected the mirror function between the pairs of neocortical neurons coupled by STDP.
Collapse
Affiliation(s)
- Gabriela Antunes
- Department of Physics, Faculdade de Filosofia, Ciencias e Letras de Ribeirao Preto, Universidade de Sao Paulo, Ribeirao Preto, SP, Brazil
| | | | - Fabio M. Simoes de Souza
- Center for Mathematics, Computation and Cognition, Federal University of ABC, Sao Bernardo do Campo, SP, Brazil
| |
Collapse
|